1
|
Han X, Wang X, Yan J, Song P, Wang Y, Kang Y, Rauf A, Zhang H. Multifunctional biosynthesized magnetosome for multimodal imaging and combined therapy of tumor. Mater Today Bio 2025; 30:101429. [PMID: 39839492 PMCID: PMC11750283 DOI: 10.1016/j.mtbio.2024.101429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
The large recruitment of tumor-associated macrophages and low exposure of tumor-associated antigens in tumor microenvironment have severely suppress the efficacy of anti-tumor immunotherapy. Herein, biosynthesized magnetosome (Mag) from bacteria was loaded with photothermal/photodynamic agent/near infrared (NIR) fluorescence dye (IR780) and further modified with lipid-PEG-c(RGDyK) through biomembrane, forming IMagRGD for fluorescence imaging, magnetic resonance imaging, immunotherapy and photodynamic/photothermal therapy. After intravenous injection into B16F10 tumor-bearing mice, IMagRGD could efficiently accumulate in tumor tissues based on near infrared (NIR) fluorescence and magnetic resonance dual-modality imaging, and repolarize tumor-associated macrophages (TAMs) from M2 phenotype to M1 phenotype, significantly improving the effect of tumor immunotherapy. Moreover, photothermal and photodynamic effect of IR780 could kill tumor cells and elicit immunogenic cell death to mediate anti-tumor immunity, promoting dendritic cells (DCs) maturation and then activating specific effector T cells to further eliminate tumor cells. This study provides a new approach for reversing the activity of tumor immunosuppressive microenvironment and strengthening the efficiency of tumor photoimmunotherapy.
Collapse
Affiliation(s)
- Xiaoqing Han
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Xingbo Wang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiao Yan
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Panpan Song
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yanjing Wang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yaqing Kang
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Ambar, 23430, Pakistan
| | - Haiyuan Zhang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Biomedical Engineering & The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
2
|
Zhu Z, Ouyang Q, Zhou L, Fan C, Zheng M, Nezamzadeh-Ejhieh A, Yuan H, Peng Y, Liu J. Current status and prospects of detection of breast cancer by MOFs platform. J Mol Struct 2025; 1321:139797. [DOI: 10.1016/j.molstruc.2024.139797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Zhang L, Yang C, Li J, Wang L, Zhang Z, Su M, Jiang M, Yang Q, Fu T, He L, Tan W. Efficient and Rapid Enrichment of Extracellular Vesicles Using DNA Nanotechnology-Enabled Synthetic Nano-Glue. Anal Chem 2025; 97:1557-1564. [PMID: 39807532 DOI: 10.1021/acs.analchem.4c03842] [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
Swift and efficient enrichment and isolation of extracellular vesicles (EVs) are crucial for enhancing precise disease diagnostics and therapeutic strategies, as well as elucidating the complex biological roles of EVs. Conventional methods of isolating EVs are often marred by lengthy and laborious processes. In this study, we introduce an innovative approach to enrich and isolate EVs by leveraging the capabilities of DNA nanotechnology. We have developed a novel multivalent cholesterol-modified paranemic crossover DNA (PX-DNA-chol) construct, which is a four-stranded DNA structure containing adjacent double helices intertwined with their local helix axes parallel and serves as an effective synthetic nano-glue. This construct promotes the rapid coalescence of nanoscale EVs into clusters of micrometer scale, thereby streamlining their enrichment. Utilizing a conventional low-speed centrifuge, this intriguing methodology achieves a rapid concentration of EVs within minutes, bypassing the laborious and high-speed centrifugation steps typically required. The quality of EVs isolated by our technique is comparable to that obtained through ultracentrifugation methods. Given these advancements, our PX-DNA-chol-facilitated EVs enrichment protocol is poised to advance the field of EVs research, providing a robust and accessible tool for in-depth studies of EVs.
Collapse
Affiliation(s)
- Lizhuan Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Cai Yang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Juncai Li
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Lu Wang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ziwen Zhang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Minhui Su
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Mengyuan Jiang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Qiuxia Yang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ting Fu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Lei He
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Weihong Tan
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
4
|
Feng H, Gao H, Chen J, Zhao R, Huang Y. Emerging phospholipid-targeted affinity materials for extracellular vesicle isolation and molecular profiling. J Chromatogr A 2025; 1741:465639. [PMID: 39742681 DOI: 10.1016/j.chroma.2024.465639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/21/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Extracellular vesicles (EVs) carrying lipids, proteins, nucleic acids and small molecular metabolites have emerged as an attractive paradigm for understanding and interfering physiological and pathological processes. To this end, selective and efficient separation approaches are highly demanded to obtain target EVs from complicated biosamples. With increasing knowledges on EV lipids, recent years have witnessed rapid advances of phospholipid-targeted affinity materials and platforms for high-performance isolation and analysis of EVs. In view of this, this review is contributed to introduce recent progresses in lipid membrane-targeted affinity strategies for EV isolation and molecular detection in biofluids. Affinity ligands including lipids, peptides, small molecules and aptamers and their molecular interactions with lipids are discussed in focus. The design, construction and mechanism of actions of affinity interfaces are summarized. The EV separation performances in complex biosamples and downstream proteomic, lipidomic and metabolic profiling are introduced. Finally, the perspectives and challenges for the development of next-generation phospholipid-targeted EV separation platforms are discussed.
Collapse
Affiliation(s)
- Huixia Feng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Gao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian Chen
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemistry, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
5
|
Khristov V, Weber SR, Caton-Darby M, Campbell G, Sundstrom JM. Diagnostic and Therapeutic Utility of Extracellular Vesicles in Ocular Disease. Int J Mol Sci 2025; 26:836. [PMID: 39859553 PMCID: PMC11765869 DOI: 10.3390/ijms26020836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer particles released by virtually all cells, with prominent roles in both physiological and pathological processes. The size, number, and molecular composition of released EVs correlate to the cells of origin, modulated by the cell's environment and pathologic state. The proteins, DNA, RNA, and protein cargo carried by EVs are protected by degradation, with a prominent role in targeted intercellular signaling. These properties make EVs salient targets as both carriers of biomarkers and potential therapeutic delivery vehicles. The majority of EV research has focused on blood, urine, saliva, and cerebrospinal fluid due to easy accessibility. EVs have also been identified and studied in all ocular biofluids, including the vitreous humor, the aqueous humor, and the tear film, and the study of EVs in ocular disease is a new, promising, and underexplored direction with unique challenges and considerations. This review covers recent advances in the diagnostic and therapeutic use of ocular EVs, with a focus on human applications and key preceding in vitro and in vivo animal studies. We also discuss future directions based on the study of EVs in other organ systems and disease sates.
Collapse
Affiliation(s)
- Vladimir Khristov
- Penn State Hershey College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (V.K.); (G.C.)
| | - Sarah R. Weber
- Department of Ophthalmology, Penn State University, Hershey, PA 17033, USA; (S.R.W.); (M.C.-D.)
| | - Mireille Caton-Darby
- Department of Ophthalmology, Penn State University, Hershey, PA 17033, USA; (S.R.W.); (M.C.-D.)
| | - Gregory Campbell
- Penn State Hershey College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (V.K.); (G.C.)
| | - Jeffrey M. Sundstrom
- Department of Ophthalmology, Penn State University, Hershey, PA 17033, USA; (S.R.W.); (M.C.-D.)
| |
Collapse
|
6
|
Yang L, Zhang J, Zhang J, Hou T, Gao Q, Liu X, Li F. CRISPR-Based Homogeneous Electrochemical Strategy for Near-Zero Background Detection of Breast Cancer Extracellular Vesicles via Fluidity-Enhanced Magnetic Capture Nanoprobe. Anal Chem 2025. [PMID: 39829104 DOI: 10.1021/acs.analchem.4c05181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Precise identification and analysis of multiple protein biomarkers on the surface of breast cancer cell-derived extracellular vesicles (BC-EVs) are of great significance for noninvasive diagnosis of the breast cancer subtypes, but it remains a major challenge owing to their high heterogeneity and low abundance. Herein, we established a CRISPR-based homogeneous electrochemical strategy for near-zero background and ultrasensitive detection of BC-EVs. To realize the high-performance capture and isolation of BC-EVs, fluidity-enhanced magnetic nanoprobes were facilely prepared. After capturing BC-EVs, the AND logic gate-based catalytic hairpin assembly (CHA) and the trans-cleavage activity of CRISPR-Cas12a against the magnetic signal nanoprobes were triggered successively, generating a significant electrochemical signal. Notably, the as-developed metal-mediated magnetic signal nanoprobes could efficiently decrease the background signal by magnetic separation, endowing the method with a high signal-to-noise ratio. Consequently, by ingeniously integrating DNA logic gate-based CRISPR-CHA signal amplification with dual magnetic nanoprobes in a homogeneous electrochemical strategy, precise identification and ultrasensitive detection of BC-EVs was successfully achieved through simultaneous and specific recognition of dual protein markers on the BC-EVs surface. More importantly, this approach could effectively discriminate specific subgroups of BC-EVs in clinical serum samples, which may provide great opportunities for the accurate diagnosis and prognosis evaluation of breast cancer in a noninvasive manner.
Collapse
Affiliation(s)
- Limin Yang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Jingang Zhang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Jing Zhang
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao 266011, People's Republic of China
| | - Ting Hou
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Qian Gao
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Xiaojuan Liu
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Feng Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| |
Collapse
|
7
|
Qian J, Xia J, Chiang S, Liu JF, Li K, Li F, Wei F, Aziz M, Kim Y, Go V, Morizio J, Zhong R, He Y, Yang K, Yang OO, Wong DTW, Lee LP, Huang TJ. Rapid and comprehensive detection of viral antibodies and nucleic acids via an acoustofluidic integrated molecular diagnostics chip: AIMDx. SCIENCE ADVANCES 2025; 11:eadt5464. [PMID: 39813350 PMCID: PMC11734728 DOI: 10.1126/sciadv.adt5464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Precise and rapid disease detection is critical for controlling infectious diseases like COVID-19. Current technologies struggle to simultaneously identify viral RNAs and host immune antibodies due to limited integration of sample preparation and detection. Here, we present acoustofluidic integrated molecular diagnostics (AIMDx) on a chip, a platform enabling high-speed, sensitive detection of viral immunoglobulins [immunoglobulin A (IgA), IgG, and IgM] and nucleic acids. AIMDx uses acoustic vortexes and Gor'kov potential wells at a 1/10,000 subwavelength scale for concurrent isolation of viruses and antibodies while excluding cells, bacteria, and large (>200 nanometers) vesicles from saliva samples. The chip facilitates on-chip viral RNA enrichment, lysis in 2 minutes, and detection via transcription loop-mediated isothermal amplification, alongside electrochemical sensing of antibodies, including mucin-masked IgA. AIMDx achieved nearly 100% recovery of viruses and antibodies, a 32-fold RNA detection improvement, and an immunity marker sensitivity of 15.6 picograms per milliliter. This breakthrough provides a transformative tool for multiplex diagnostics, enhancing early infectious disease detection.
Collapse
Affiliation(s)
- Jiao Qian
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Jianping Xia
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Samantha Chiang
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jessica F. Liu
- Department of Anesthesiology, Duke University, Durham, NC 27710, USA
| | - Ke Li
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Feng Li
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Fang Wei
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Mohammad Aziz
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Vinson Go
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 27710, USA
| | - James Morizio
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 27710, USA
| | - Ruoyu Zhong
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Ye He
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Kaichun Yang
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David T. W. Wong
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Luke P. Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea
| | - Tony Jun Huang
- Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, NC 27708, USA
| |
Collapse
|
8
|
Wang L, Gong Z, Wang M, Liang YZ, Zhao J, Xie Q, Wu XW, Li QY, Zhang C, Ma LY, Zheng SY, Jiang M, Yu X, Xu L. Rapid and unbiased enrichment of extracellular vesicles via a meticulously engineered peptide. Bioact Mater 2025; 43:292-304. [PMID: 39399836 PMCID: PMC11470464 DOI: 10.1016/j.bioactmat.2024.09.023] [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: 05/16/2024] [Revised: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024] Open
Abstract
Extracellular vesicles (EVs) have garnered significant attention in biomedical applications. However, the rapid, efficient, and unbiased separation of EVs from complex biological fluids remains a challenge due to their heterogeneity and low abundance in biofluids. Herein, we report a novel approach to reconfigure and modify an artificial insertion peptide for the unbiased and rapid isolation of EVs in 20 min with ∼80% recovery in neutral conditions. Moreover, the approach demonstrates exceptional anti-interference capability and achieves a high purity of EVs comparable to standard ultracentrifugation and other methods. Importantly, the isolated EVs could be directly applied for downstream protein and nucleic acid analyses, including proteomics analysis, exome sequencing analysis, as well as the detection of both epidermal growth factor receptor (EGFR) and V-Ki-ras2 Kirsten Rat Sarcoma Viral Oncogene Homologue (KRAS) gene mutation in clinical plasma samples. Our approach offers great possibilities for utilizing EVs in liquid biopsy, as well as in various other biomedical applications.
Collapse
Affiliation(s)
- Le Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhou Gong
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Innovation Academy for Precision Measurement Science and Technology Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ming Wang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi-Zhong Liang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qi Xie
- College of Horticulture and Forestry Sciences, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiao-Wei Wu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical Collage of Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin-Ying Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cong Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li-Yun Ma
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Si-Yang Zheng
- Department of Electrical Engineering and Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, United States
| | - Ming Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xu Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
9
|
Wang Y, Chen H, Xie L, Liu J, Zhang L, Yu J. Swarm Autonomy: From Agent Functionalization to Machine Intelligence. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2312956. [PMID: 38653192 PMCID: PMC11733729 DOI: 10.1002/adma.202312956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Swarm behaviors are common in nature, where individual organisms collaborate via perception, communication, and adaptation. Emulating these dynamics, large groups of active agents can self-organize through localized interactions, giving rise to complex swarm behaviors, which exhibit potential for applications across various domains. This review presents a comprehensive summary and perspective of synthetic swarms, to bridge the gap between the microscale individual agents and potential applications of synthetic swarms. It is begun by examining active agents, the fundamental units of synthetic swarms, to understand the origins of their motility and functionality in the presence of external stimuli. Then inter-agent communications and agent-environment communications that contribute to the swarm generation are summarized. Furthermore, the swarm behaviors reported to date and the emergence of machine intelligence within these behaviors are reviewed. Eventually, the applications enabled by distinct synthetic swarms are summarized. By discussing the emergent machine intelligence in swarm behaviors, insights are offered into the design and deployment of autonomous synthetic swarms for real-world applications.
Collapse
Affiliation(s)
- Yibin Wang
- School of Science and EngineeringThe Chinese University of Hong KongShenzhen518172China
- Shenzhen Institute of Artificial Intelligence and Robotics for SocietyShenzhen518172China
| | - Hui Chen
- School of Science and EngineeringThe Chinese University of Hong KongShenzhen518172China
- Shenzhen Institute of Artificial Intelligence and Robotics for SocietyShenzhen518172China
| | - Leiming Xie
- School of Science and EngineeringThe Chinese University of Hong KongShenzhen518172China
- Shenzhen Institute of Artificial Intelligence and Robotics for SocietyShenzhen518172China
| | - Jinbo Liu
- School of Science and EngineeringThe Chinese University of Hong KongShenzhen518172China
- Shenzhen Institute of Artificial Intelligence and Robotics for SocietyShenzhen518172China
| | - Li Zhang
- Department of Mechanical and Automation EngineeringThe Chinese University of Hong KongHong Kong999077China
| | - Jiangfan Yu
- School of Science and EngineeringThe Chinese University of Hong KongShenzhen518172China
- Shenzhen Institute of Artificial Intelligence and Robotics for SocietyShenzhen518172China
| |
Collapse
|
10
|
Cipa J, Endzelins E, Abols A, Romanchikova N, Line A, Jenster GW, Mozolevskis G, Rimsa R. Elucidating Extracellular Vesicle Isolation Kinetics via an Integrated Off-Stoichiometry Thiol-Ene and Cyclic Olefin Copolymer Microfluidic Device. Polymers (Basel) 2024; 16:3579. [PMID: 39771431 PMCID: PMC11678796 DOI: 10.3390/polym16243579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Extracellular vesicles (EVs) are promising biomarkers for diagnosing complex diseases such as cancer and neurodegenerative disorders. Yet, their clinical application is hindered by challenges in isolating cancer-derived EVs efficiently due to their broad size distribution in biological samples. This study introduces a microfluidic device fabricated using off-stoichiometry thiol-ene and cyclic olefin copolymer, addressing the absorption limitations of polydimethylsiloxane (PDMS). The device streamlines a standard laboratory assay into a semi-automated microfluidic chip, integrating sample mixing and magnetic particle separation. Using the microfluidic device, the binding kinetics between EVs and anti-CD9 nanobodies were measured for the first time. Based on the binding kinetics, already after 10 min the EV capture was saturated and comparable to standard laboratory assays, offering a faster alternative to antibody-based immunomagnetic protocols. Furthermore, this study reveals the binding kinetics of EVs to anti-CD9 nanobodies for the first time. Our findings demonstrate the potential of the microfluidic device to enhance clinical diagnostics by offering speed and reducing manual labor without compromising accuracy.
Collapse
Affiliation(s)
- Janis Cipa
- Institute of Solid State Physics, University of Latvia, 8 Kengaraga Str., LV-1063 Riga, Latvia; (J.C.); (G.M.)
- Cellbox Labs LLC, 8 Kengaraga Str., LV-1063 Riga, Latvia;
| | - Edgars Endzelins
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, LV-1067 Riga, Latvia; (E.E.); (N.R.); (A.L.)
| | - Arturs Abols
- Cellbox Labs LLC, 8 Kengaraga Str., LV-1063 Riga, Latvia;
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, LV-1067 Riga, Latvia; (E.E.); (N.R.); (A.L.)
| | - Nadezda Romanchikova
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, LV-1067 Riga, Latvia; (E.E.); (N.R.); (A.L.)
| | - Aija Line
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, LV-1067 Riga, Latvia; (E.E.); (N.R.); (A.L.)
| | - Guido W. Jenster
- Department of Urology, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Gatis Mozolevskis
- Institute of Solid State Physics, University of Latvia, 8 Kengaraga Str., LV-1063 Riga, Latvia; (J.C.); (G.M.)
- Cellbox Labs LLC, 8 Kengaraga Str., LV-1063 Riga, Latvia;
| | - Roberts Rimsa
- Institute of Solid State Physics, University of Latvia, 8 Kengaraga Str., LV-1063 Riga, Latvia; (J.C.); (G.M.)
- Cellbox Labs LLC, 8 Kengaraga Str., LV-1063 Riga, Latvia;
| |
Collapse
|
11
|
Yang C, Li N, Chen H, Zhang M, Chen Y, Zhang X, Huang S, Sun N, Deng C. In Situ Array Microextraction and Metabolic Profiling of Small Extracellular Vesicles to Guide and Monitor Maternal Delivery. SMALL METHODS 2024; 8:e2400261. [PMID: 38837641 DOI: 10.1002/smtd.202400261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/27/2024] [Indexed: 06/07/2024]
Abstract
The advantages of small extracellular vesicles (sEV) in disease management have become increasingly prominent, with the main challenge lying in meeting the demands of large-scale extraction and high-throughput analysis, a crucial aspect in the realm of precision medicine. To overcome this challenge, an engineered on-plate aptamer array (16×24 spots) is developed for continuous scale-up microextraction of plasma sEV and their in situ metabolic analysis using mass spectrometry. With this integrated array strategy, metabolic profiles of sEV are acquired from the plasma of 274 antenatal or postpartum women, reducing analysis time by half (7.5 h) and sample volume by 95% (only 0.125 µL usage) compared to the traditional suspension method. Moreover, using machine learning algorithms on sEV metabolic profiles, a risk score system is constructed that accurately assesses the need for epidural analgesia during childbirth and the likelihood of post-administration fever. The system, based on admission samples, achieves an impressive 94% accuracy. Furthermore, post-administration fever can be identified from delivery samples, reaching an overall accuracy rate of 88%. This work offers real-time monitoring of the childbirth process that can provide timely guidance for maternal delivery, underscoring the significance of sEV detection in large-scale clinical samples for medicine innovation and advancement.
Collapse
Affiliation(s)
- Chenyu Yang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Ning Li
- Department of Anesthesia, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200090, China
| | - Haolin Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Man Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Yijie Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Xiangmin Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Shaoqiang Huang
- Department of Anesthesia, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, 200090, China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China
| |
Collapse
|
12
|
Xiao J, Ding J, Sun C, Liu D, Gao H, Liu Y, Lu Y, Gao X. Simultaneous Detection of Clenbuterol and Higenamine in Urine Samples Using Interference-Free SERS Tags Combined with Magnetic Separation. ACS Sens 2024; 9:5394-5404. [PMID: 39383499 DOI: 10.1021/acssensors.4c01623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
Sports doping remains a significant challenge in competitive sports. Given that urine analysis is the standard for detecting doping, developing rapid, sensitive, accurate, and high-throughput methods for stimulant detection in urine is crucial. Surface-enhanced Raman scattering (SERS) tag-based immunoassays have emerged as powerful analytical tools known for their high sensitivity and specificity, holding particular promise for stimulant detection in urine samples. However, both the Raman signals of typical SERS tags and sample matrices are within the Raman fingerprint region (<1800 cm-1), which could lead to spectrum overlap, potentially reducing detection accuracy and sensitivity. By recognizing this, we designed a competitive immunoassay that integrates two types of zero-background SERS tags and magnetic separation. These innovative SERS tags exhibit distinctive Raman peaks within the Raman-silent region (1800-2800 cm-1), effectively mitigating potential spectrum overlap with background sample signals. Moreover, magnetic separation not only enhances operational simplicity but also improves the system's anti-interference capability. Using clenbuterol (CL) and higenamine (HM) as model targets, the SERS-based competitive immunoassay demonstrated sensitive detection of individual CL or HM standards, with limits of detection (LODs) of 0.87 and 0.71 pg/mL, respectively. In multiplex mode, CL and HM can be simultaneously detected with LODs of 1.0 and 0.81 pg/mL, respectively. Furthermore, the recovery rates in urine samples ranged from 83 to 116% (relative standard deviation, RSD ≤ 6.4%) for CL and from 82 to 103% (RSD ≤ 5.1%) for HM, further confirming the reliability of the SERS-based immunoassay for practical applications.
Collapse
Affiliation(s)
- Jinru Xiao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jiacheng Ding
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Chenyang Sun
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Hongmei Gao
- Department of Intensive Care Unit, Key Laboratory for Critical Care Medicine of the Ministry of Health, Emergency Medicine Research Institute, Tianjin First Center Hospital, Nankai University, Tianjin 300192, China
| | - Yaqing Liu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yang Lu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xia Gao
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
13
|
Zhang G, Huang X, Gong Y, Ding Y, Wang H, Zhang H, Wu L, Su R, Yang C, Zhu Z. Fingerprint Profiling of Glycans on Extracellular Vesicles via Lectin-Induced Aggregation Strategy for Precise Cancer Diagnostics. J Am Chem Soc 2024; 146:29053-29063. [PMID: 39235449 DOI: 10.1021/jacs.4c10390] [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: 09/06/2024]
Abstract
Extracellular vesicles (EVs) harbor abundant glycans that mediate various functions, such as intercellular communication and disease advancement, which play significant roles in disease progression. However, the presence of EV heterogeneity in body fluids and the complex nature of the glycan structures have posed challenges for the detection of EV glycans. In this study, we provide a streamlined method integrated, membrane-specific separation with lectin-induced aggregation strategy (MESSAGE), for multiplexed profiling of EV glycans. By leveraging a rationally designed lectin-induced aggregation strategy, the expression of EV glycans is converted to size-based signals. With the assistance learning machine algorithms, the MESSAGE strategy with high sensitivity, specificity, and simplicity can be used for early cancer diagnosis and classification, as well as monitoring cancer metastasis via 20 μL plasma sample within 2 h. Furthermore, our platform holds promise for advancing the field of EV-based liquid biopsy for clinical applications, opening new possibilities for the profiling of EV glycan signatures in various disease states.
Collapse
Affiliation(s)
- Guihua Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaodan Huang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yanli Gong
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yue Ding
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hua Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Huimin Zhang
- Innovation Laboratory for Sciences, Technologies of Energy Materials of Fujian Province, Xiamen 361000, China
| | - Lingling Wu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Rui Su
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Chaoyong Yang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, 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
| | - Zhi Zhu
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
14
|
Zhang J, Gao Z, Xiao W, Jin N, Zeng J, Wang F, Jin X, Dong L, Lin J, Gu J, Wang C. A simplified and efficient extracellular vesicle-based proteomics strategy for early diagnosis of colorectal cancer. Chem Sci 2024:d4sc05518g. [PMID: 39421202 PMCID: PMC11480824 DOI: 10.1039/d4sc05518g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related death worldwide and an effective screening strategy for diagnosis of early-stage CRC is highly desired. Although extracellular vesicles (EVs) are expected to become some of the most promising tools for liquid biopsy of early disease diagnosis, the existing EV-based proteomics methods for practical application in clinical samples are limited by technical challenges in high-throughput isolation and detection of EVs. In the current study, we have developed a simplified and efficient EV-based proteomics strategy for early diagnosis of CRC. DSPE-functionalized beads were specifically designed that enabled direct capture of EVs from plasma samples in 10 minutes with good reproducibility and comprehensive proteome coverage. The single-pot, solid-phase-enhanced sample-preparation (SP3) technology was then combined with data-independent acquisition mass spectrometry (DIA-MS) for in-depth analysis and quantification of EV proteomes. From a cohort with 30 individuals including 11 healthy controls, 8 patients with adenomatous polyp and 11 patients with early-stage CRC, our streamlined workflow reproducibly quantified over 800 proteins from their plasma-derived EV samples, from which dysregulated protein signatures for molecular diagnosis of CRC were revealed. We selected a panel of 10 protein markers to train a machine learning (ML) model, which resulted in accurate prediction of polyp and early-stage CRC in an independent and single-blind validation cohort with excellent diagnostic ability of 89.3% accuracy. Our simplified and efficient clinical proteomic strategy will serve as a valuable tool for fast, accurate, and cost-effective diagnosis of CRC that can be easily extended to other disease samples for discovery of unique EV-based biomarkers.
Collapse
Affiliation(s)
- Jin Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing China
| | - Zhaoya Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital Beijing China
- Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center Beijing China
| | - Weidi Xiao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies Chengdu China
| | - Ningxin Jin
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing China
| | - Jiaming Zeng
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies Chengdu China
| | - Fengzhang Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing China
| | - Xiaowei Jin
- Department of Gastroenterology, Peking University Shougang Hospital Beijing China
| | - Liguang Dong
- Center for Health Care Management, Peking University Shougang Hospital Beijing China
| | - Jian Lin
- Department of Pharmacy, NMPA Key Laboratory for Research and Evaluation of Generic Drugs, Peking University Third Hospital Cancer Center, Peking University Third Hospital Beijing China
- Synthetic and Functional Biomolecules Center, Peking University Beijing China
| | - Jin Gu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital Beijing China
- Center for Precision Diagnosis and Treatment of Colorectal Cancer and Inflammatory Disease, Peking University Health Science Center Beijing China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute Beijing China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University Beijing China
| | - Chu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies Chengdu China
- Synthetic and Functional Biomolecules Center, Peking University Beijing China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University Beijing China
| |
Collapse
|
15
|
Dlugolecka M, Czystowska-Kuzmicz M. Factors to consider before choosing EV labeling method for fluorescence-based techniques. Front Bioeng Biotechnol 2024; 12:1479516. [PMID: 39359260 PMCID: PMC11445045 DOI: 10.3389/fbioe.2024.1479516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
A well-designed fluorescence-based analysis of extracellular vesicles (EV) can provide insights into the size, morphology, and biological function of EVs, which can be used in medical applications. Fluorescent nanoparticle tracking analysis with appropriate controls can provide reliable data for size and concentration measurements, while nanoscale flow cytometry is the most appropriate tool for characterizing molecular cargoes. Label selection is a crucial element in all fluorescence methods. The most comprehensive data can be obtained if several labeling approaches for a given marker are used, as they would provide complementary information about EV populations and interactions with the cells. In all EV-related experiments, the influence of lipoproteins and protein corona on the results should be considered. By reviewing and considering all the factors affecting EV labeling methods used in fluorescence-based techniques, we can assert that the data will provide as accurate as possible information about true EV biology and offer precise, clinically applicable information for future EV-based diagnostic or therapeutic applications.
Collapse
|
16
|
Zhu N, Yu Y, An D, Zeng Y, Kang K, Yi Q, Wu Y. Harnessing Spatiotemporal-Specific Tumorous Exosome Dynamics: Ultra-Sensitive Lanthanide Luminescence Detection Strategy Enabled by Exosomal Membrane Engineering for Melanoma Immunotherapy Monitoring. ACS APPLIED MATERIALS & INTERFACES 2024; 16:49164-49175. [PMID: 39252418 DOI: 10.1021/acsami.4c10804] [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: 09/11/2024]
Abstract
Focused on the newly secreted tumorous exosomes during melanoma immunotherapy, this work has pioneered an ultra-sensitive spatiotemporal-specific exosome detection strategy, leveraging advanced exosomal membrane engineering techniques. The proposed strategy harnesses the power of amplified lanthanide luminescence signals on these exosomes, enabling precise and real-time monitoring of the efficacy of melanoma immunotherapy. The methodology comprises two pivotal steps. Initially, Ac4ManNAz-associated metabolic labeling is employed to evolve azide groups onto the membranes of newly secreted exosomes with remarkable selectivity. These azide groups serve as versatile clickable artificial tags, enabling the precise identification of melanoma exosomes emerging during immunotherapy. Subsequently, lanthanide-nanoparticle-functionalized polymer chains are controllably grafted onto the exosome surfaces through click chemistry and in situ Fenton-RAFT polymerization, serving as robust signal amplifiers. When integrated with time-resolved fluorescence detection, this strategy yields detection signals with an exceptionally high signal-to-noise ratio, enabling ultra-sensitive detection of PD-L1 antigen expression levels on the spatiotemporal-specific exosomes. The detection strategy boasts a wide linear concentration range spanning from 1.7 × 104 to 1.7 × 109 particles/mL, with a remarkable theoretical detection limit of 1.28 × 103 particles/mL. The remarkable enhancements in detection sensitivity and accuracy facilitate the evaluation of the efficacy of immunotherapeutic interventions in the mouse B16 melanoma model, notably revealing a substantial disparity in PD-L1 levels between immunotherapy-treated and untreated groups (P < 0.01) and further emphasizing the cumulative therapeutic effect that intensifies with repeated treatments (P < 0.001).
Collapse
Affiliation(s)
- Nanhang Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Yue Yu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Di An
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Yating Zeng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Ke Kang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| |
Collapse
|
17
|
Chen C, Pan X, Sun M, Wang J, Su X, Zhang T, Chen Y, Wu D, Li J, Wu S, Yan X. Phospholipid-Anchored Ligand Conjugation on Extracellular Vesicles for Enhanced Cancer Targeting. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310712. [PMID: 38733222 DOI: 10.1002/smll.202310712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/27/2024] [Indexed: 05/13/2024]
Abstract
Extracellular vesicles (EVs) are recognized as potential candidates for next-generation drug delivery systems. However, the inherent cancer-targeting efficiency is unsatisfactory, necessitating surface modification to attach cell-binding ligands. By utilizing phospholipase D from Streptomyces in combination with maleimide-containing primary alcohol, the authors successfully anchored ligands onto milk-derived EVs (mEVs), overcoming the issues of ligand leakage or functional alteration seen in traditional methods. Quantitative nano-flow cytometry demonstrated that over 90% of mEVs are effectively modified with hundreds to thousands of ligands. The resulting mEV formulations exhibited remarkable long-term stability in conjugation proportion, ligand number, size distribution, and particle concentration, even after months of storage. It is further shown that conjugating transferrin onto mEVs significantly enhanced cellular uptake and induced pronounced cytotoxic effects when loaded with paclitaxel. Overall, this study presents a highly efficient, stable, cost-effective, and scalable ligand conjugation approach, offering a promising strategy for targeted drug delivery of EVs.
Collapse
Affiliation(s)
- Chaoxiang Chen
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Xueping Pan
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Mengdi Sun
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Jialin Wang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Xueqi Su
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Tianyu Zhang
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Yulei Chen
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Daren Wu
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Jian Li
- Department of Biological Engineering, College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, 361021, China
| | - Shuqi Wu
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an, 710072, 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 Engineering, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
18
|
Weerakkody JS, Tseng T, Topper M, Thoduvayil S, Radhakrishnan A, Pincet F, Kyriakides TR, Gunasekara RW, Ramakrishnan S. Photosensitive Nanoprobes for Rapid Isolation and Size-Specific Enrichment of Synthetic and Extracellular Vesicle Subpopulations. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2400390. [PMID: 39372670 PMCID: PMC11452007 DOI: 10.1002/adfm.202400390] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Indexed: 10/08/2024]
Abstract
The biggest challenge in current isolation methods for lipid bilayer-encapsulated vesicles, such as exosomes, secretory, and synthetic vesicles, lies in the absence of a unified approach that seamlessly delivers high purity, yield, and scalability for large-scale applications. To address this gap, we have developed an innovative method that utilizes photosensitive lipid nanoprobes specifically designed for efficient isolation of vesicles and sorting them into subpopulations based on size. The photosensitive component in the probe undergoes cleavage upon exposure to light, facilitating the release of vesicles in their near-native form. We demonstrate that our method provides superior capability in isolating extracellular vesicles from complex biological media and separating them into size-based subpopulations within 1 hour, achieving more efficiency and purity than ultracentrifugation. Furthermore, this method's cost-effectiveness and rapid enrichment of the vesicles align with demands for large-scale isolation and downstream analyses of nucleic acids and proteins. Our method opens new avenues in exploring, analyzing, and utilizing synthetic and extracellular vesicle subpopulations in various biomedical applications, including diagnostics, therapeutic delivery, and biomarker discovery.
Collapse
Affiliation(s)
- Jonathan S. Weerakkody
- Yale Nanobiology Institute, West Haven, CT 06516, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Tiffany Tseng
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | - Sikha Thoduvayil
- Yale Nanobiology Institute, West Haven, CT 06516, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Abhijith Radhakrishnan
- Yale Nanobiology Institute, West Haven, CT 06516, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Frederic Pincet
- Yale Nanobiology Institute, West Haven, CT 06516, USA
- Laboratoire de Physique de l’Ecole Normale Superieure, Ecole Normale Superieure (ENS), Universite Paris Sciences et Lettres (PSL), CNRS, Sorbonne Universite, Universite Paris-Cite, 75005 Paris, France
| | - Themis R. Kyriakides
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Roshan W. Gunasekara
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Sathish Ramakrishnan
- Yale Nanobiology Institute, West Haven, CT 06516, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
19
|
Xia J, Wang Z, Becker R, Li F, Wei F, Yang S, Rich J, Li K, Rufo J, Qian J, Yang K, Chen C, Gu Y, Zhong R, Lee PJ, Wong DTW, Lee LP, Huang TJ. Acoustofluidic Virus Isolation via Bessel Beam Excitation Separation Technology. ACS NANO 2024; 18:22596-22607. [PMID: 39132820 DOI: 10.1021/acsnano.4c09692] [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: 08/13/2024]
Abstract
The isolation of viruses from complex biological samples is essential for creating sensitive bioassays that assess the efficacy and safety of viral therapeutics and vaccines, which have played a critical role during the COVID-19 pandemic. However, existing methods of viral isolation are time-consuming and labor-intensive due to the multiple processing steps required, resulting in low yields. Here, we introduce the rapid, efficient, and high-resolution acoustofluidic isolation of viruses from complex biological samples via Bessel beam excitation separation technology (BEST). BEST isolates viruses by utilizing the nondiffractive and self-healing properties of 2D, in-plane acoustic Bessel beams to continuously separate cell-free viruses from biofluids, with high throughput and high viral RNA yield. By tuning the acoustic parameters, the cutoff size of isolated viruses can be easily adjusted to perform dynamic, size-selective virus isolation while simultaneously trapping larger particles and separating smaller particles and contaminants from the sample, achieving high-precision isolation of the target virus. BEST was used to isolate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from human saliva samples and Moloney Murine Leukemia Virus from cell culture media, demonstrating its potential use in both practical diagnostic applications and fundamental virology research. With high separation resolution, high yield, and high purity, BEST is a powerful tool for rapidly and efficiently isolating viruses. It has the potential to play an important role in the development of next-generation viral diagnostics, therapeutics, and vaccines.
Collapse
Affiliation(s)
- Jianping Xia
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Zeyu Wang
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Ryan Becker
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Feng Li
- School of Dentistry, University of California, Los Angeles, California 90095, United States
| | - Fang Wei
- School of Dentistry, University of California, Los Angeles, California 90095, United States
| | - Shujie Yang
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Joseph Rich
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Ke Li
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Joseph Rufo
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Jiao Qian
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Kaichun Yang
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Chuyi Chen
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Yuyang Gu
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Ruoyu Zhong
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| | - Patty J Lee
- Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - David T W Wong
- School of Dentistry, University of California, Los Angeles, California 90095, United States
| | - Luke P Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Boston, Massachusetts 02115, United States
| | - Tony Jun Huang
- The Thomas Lord Department of Mechanical Engineering and Materials, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
20
|
Lei H, Wang H, Wang X, Xiao Z, Tian T, Cui K. Surface-enhanced Raman scattering-based identification of breast cancer progression using extracellular vesicles-derived integrin α6β4. Talanta 2024; 275:126092. [PMID: 38615459 DOI: 10.1016/j.talanta.2024.126092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Detection of progression is of great importance to breast cancer treatment and can benefit patients. Limited by current detection technologies and biomarkers, early breast cancer progression diagnosis remains challenging. Researchers have found blood extracellular vesicles (EVs)-derived integrin α6β4 directly facilitate progression in breast cancer, enabling cancer detection. However, EVs size and heterogeneity hinder protein detection, masked by abundant background EVs. Hence, novel tools for efficient detection of EVs with high selectivity and low interference are significantly desired. Here, a new silver-coated gold nanorods SERS probe, termed as Au@Ag@IDA-B/4MSTP, based on DNA aptamer was established for the detection of integrin α6β4 derived from EVs. Validation of the Au@Ag@IDA-B/4MSTP probes using cell-culture-derived EVs revealed a LOD of 23 particles/μL for EVs detection. This tool was further confirmed to mimic the real state of cancer with subcutaneous tumor model and lung metastasis model in mice. With 10 μL of blood plasma and simple Raman analysis process, the test achieved 85.7 % sensitivity and 83.3 % specificity. Moreover, our method achieves a simplified approach that expedites the detection process. These results demonstrate the good detection performance of Au@Ag@IDA-B/4MSTP probes for EVs integrin α6β4, and suggest that this non-invasive approach could be a promising tool for early detection of breast cancer progression.
Collapse
Affiliation(s)
- Haozhi Lei
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Shanghai, 200127, China
| | - Haoze Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China; College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200233, China
| | - Xiqiu Wang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zeyu Xiao
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China; Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Tian Tian
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Kai Cui
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
21
|
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] [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
|
22
|
Yang Z, Wu H, Wang Z, Bian E, Zhao B. The role and application of small extracellular vesicles in glioma. Cancer Cell Int 2024; 24:229. [PMID: 38951882 PMCID: PMC11218314 DOI: 10.1186/s12935-024-03389-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024] Open
Abstract
Small extracellular vesicles (sEVs) are cell-derived, nanometer-sized particles enclosed by a lipid bilayer. All kinds of biological molecules, including proteins, DNA fragments, RNA, lipids, and metabolites, can be selectively loaded into sEVs and transmitted to recipient cells that are near and distant. Growing shreds of evidence show the significant biological function and the clinical significance of sEVs in cancers. Numerous recent studies have validated that sEVs play an important role in tumor progression and can be utilized to diagnose, stage, grading, and monitor early tumors. In addition, sEVs have also served as drug delivery nanocarriers and cancer vaccines. Although it is still infancy, the field of basic and translational research based on sEVs has grown rapidly. In this review, we summarize the latest research on sEVs in gliomas, including their role in the malignant biological function of gliomas, and the potential of sEVs in non-invasive diagnostic and therapeutic approaches, i.e., as nanocarriers for drug or gene delivery and cancer vaccines.
Collapse
Affiliation(s)
- Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - HaoYuan Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - ZhiWei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - ErBao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China.
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China.
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China.
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China.
| |
Collapse
|
23
|
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
|
24
|
Chen M, Li J, Lin Y, Li X, Yu Y, Zhou S, Xu F, Zhang Q, Zhang H, Wang W. Recent research on material-based methods for isolation of extracellular vesicles. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3179-3191. [PMID: 38738644 DOI: 10.1039/d4ay00370e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Extracellular vesicles (EVs) are nanoparticles secreted by cells with a closed phospholipid bilayer structure, which can participate in various physiological and pathological processes and have significant clinical value in disease diagnosis, targeted therapy and prognosis assessment. EV isolation methods currently include differential ultracentrifugation, ultrafiltration, size exclusion chromatography, immunoaffinity, polymer co-precipitation and microfluidics. In addition, material-based biochemical or biophysical approaches relying on intrinsic properties of the material or its surface-modified functionalized monomers, demonstrated unique advantages in the efficient isolation of EVs. In order to provide new ideas for the subsequent development of material-based EV isolation methods, this review will focus on the principle, research status and application prospects of material-based EV isolation methods based on different material carriers and functional monomers.
Collapse
Affiliation(s)
- Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Jiaxi Li
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Yujie Lin
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Xiaowei Li
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, PR China
| | - Yuanyuan Yu
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Shenyue Zhou
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Fang Xu
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Qi Zhang
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Haiyang Zhang
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Yunxuan Building #1339 and #2103, Wenjing Road, Suzhou Industrial Park, Suzhou 215123, China.
| |
Collapse
|
25
|
Guo X, Hu F, Yong Z, Zhao S, Wan Y, Wang B, Peng N. Magnetic Nanoparticle-Based Microfluidic Platform for Automated Enrichment of High-Purity Extracellular Vesicles. Anal Chem 2024; 96:7212-7219. [PMID: 38660946 DOI: 10.1021/acs.analchem.4c00795] [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: 04/26/2024]
Abstract
Extracellular vesicles (EVs) are available in various biological fluids and have highly heterogeneous sizes, origins, contents, and functions. Rapid enrichment of high-purity EVs remains crucial for enhancing research on EVs in tumors. In this work, we present a magnetic nanoparticle-based microfluidic platform (ExoCPR) for on-chip isolation, purification, and mild recovery of EVs from cell culture supernatant and plasma within 29 min. The ExoCPR chip integrates bubble-driven micromixers and immiscible filtration assisted by surface tension (IFAST) technology. The bubble-driven micromixer enhances the mixing between immunomagnetic beads and EVs, eliminating the need for manual pipetting or off-chip oscillatory incubation. The high-purity EVs were obtained after passing through the immiscible phase interface where hydrophilic or hydrophobic impurities nonspecifically bound to SIMI were removed. The ExoCPR chip had a capture efficiency of 75.8% and a release efficiency of 62.7% for model EVs. We also demonstrated the powerful performance of the ExoCPR in isolating EVs from biological samples (>90% purity). This chip was further employed in clinical plasma samples and showed that the number of GPC3-positive EVs isolated from hepatocellular carcinoma patients was significantly higher than that of healthy individuals. This ExoCPR chip may provide a promising tool for EV-based liquid biopsy and other fundamental research.
Collapse
Affiliation(s)
- Xiaoniu Guo
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Zhang Yong
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Shuhao Zhao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Yong Wan
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Bingqing Wang
- Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| | - Niancai Peng
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| |
Collapse
|
26
|
Tong Z, Yang D, Shen C, Li C, Xu X, Li Q, Wu Z, Ma H, Chen F, Mao H. Rapid automated extracellular vesicle isolation and miRNA preparation on a cost-effective digital microfluidic platform. Anal Chim Acta 2024; 1296:342337. [PMID: 38401929 DOI: 10.1016/j.aca.2024.342337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/26/2024]
Abstract
As a prerequisite for extracellular vesicle (EV) -based studies and diagnosis, effective isolation, enrichment and retrieval of EV biomarkers are crucial to subsequent analyses, such as miRNA-based liquid biopsy for non-small-cell lung cancer (NSCLC). However, most conventional approaches for EV isolation suffer from lengthy procedure, high cost, and intense labor. Herein, we introduce the digital microfluidic (DMF) technology to EV pretreatment protocols and demonstrate a rapid and fully automated sample preparation platform for clinical tumor liquid biopsy. Combining a reusable DMF chip technique with a low-cost EV isolation and miRNA preparation protocol, the platform completes automated sample processing in 20-30 min, supporting immediate RT-qPCR analyses on EV-derived miRNAs (EV-miRNAs). The utility and reliability of the platform was validated via clinical sample processing for EV-miRNA detection. With 23 tumor and 20 non-tumor clinical plasma samples, we concluded that EV-miR-486-5p and miR-21-5p are effective biomarkers for NSCLC with a small sample volumn (20-40 μL). The result was consistent to that of a commercial exosome miRNA extraction kit. These results demonstrate the effectiveness of DMF in EV pretreatment for miRNA detection, providing a facile solution to EV isolation for liquid biopsy.
Collapse
Affiliation(s)
- Zhaoduo Tong
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dawei Yang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuanjie Shen
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Li
- Department of Neurosurgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xin Xu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Qiushi Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Zhenhua Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Ma
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Fuxiang Chen
- Department of Clinical Immunology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hongju Mao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
27
|
Shen X, Zhang Y, Wang D, Huang Y, Song Y, Wang S. Mediator Monomer Regulated Emulsion Interfacial Polymerization to Synthesize Nanofractal Magnetic Particles for Nucleic Acid Separation. SMALL METHODS 2024; 8:e2300531. [PMID: 37491768 DOI: 10.1002/smtd.202300531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/14/2023] [Indexed: 07/27/2023]
Abstract
Polymer-based magnetic particles have been widely used for the separation of biological samples including nucleic acids, proteins, virus, and cells. Existing magnetic particles are almost prepared by coating polymers on magnetic nanoparticles (NPs). However, this strategy usually encounters the problem of poor magnetic NPs loading capacity. Here, a series of nanofractal magnetic particles (nanoFMPs) synthesized by a strategy of mediator monomer regulated emulsion interfacial polymerization is presented, which allows effective magnetic NPs loading and show efficient nucleic acid separation performance. The mediator monomers facilitate the dispersion of magnetic NPs in internal phase to achieve higher loading, and the hydrophilic monomers use electrostatic interactions to form surface nanofractal structures with functional groups. Compared with magnetic particles without nanofractal structure, nanoFMPs exhibit a higher nucleic acid extraction capability. This strategy offers an effective and versatile way for the synthesis of nanoFMPs toward efficient separation in various fields from clinical diagnosis to food safety and environmental monitoring.
Collapse
Affiliation(s)
- Xinyi Shen
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yue Zhang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Duanda Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanling Huang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yongyang Song
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Binzhou Institute of Technology, Weiqiao-UCAS Science and Technology Park, Binzhou City, Shandong Province, 256606, P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Binzhou Institute of Technology, Weiqiao-UCAS Science and Technology Park, Binzhou City, Shandong Province, 256606, P. R. China
| |
Collapse
|
28
|
Wu Z, Cai H, Tian C, Ao Z, Jiang L, Guo F. Exploiting Sound for Emerging Applications of Extracellular Vesicles. NANO RESEARCH 2024; 17:462-475. [PMID: 38712329 PMCID: PMC11073796 DOI: 10.1007/s12274-023-5840-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/08/2024]
Abstract
Extracellular vesicles are nano- to microscale, membrane-bound particles released by cells into extracellular space, and act as carriers of biomarkers and therapeutics, holding promising potential in translational medicine. However, the challenges remain in handling and detecting extracellular vesicles for disease diagnosis as well as exploring their therapeutic capability for disease treatment. Here, we review the recent engineering and technology advances by leveraging the power of sound waves to address the challenges in diagnostic and therapeutic applications of extracellular vesicles and biomimetic nanovesicles. We first introduce the fundamental principles of sound waves for understanding different acoustic-assisted extracellular vesicle technologies. We discuss the acoustic-assisted diagnostic methods including the purification, manipulation, biosensing, and bioimaging of extracellular vesicles. Then, we summarize the recent advances in acoustically enhanced therapeutics using extracellular vesicles and biomimetic nanovesicles. Finally, we provide perspectives into current challenges and future clinical applications of the promising extracellular vesicles and biomimetic nanovesicles powered by sound.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Lei Jiang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, United States
| |
Collapse
|
29
|
Jiao Y, Gao L, Zhang T, He Z, Zheng SY, Liu W. Profiling DNA Cargos in Single Extracellular Vesicles via Hydrogel-Based Droplet Digital Multiple Displacement Amplification. Anal Chem 2024; 96:1293-1300. [PMID: 38189229 DOI: 10.1021/acs.analchem.3c04666] [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: 01/09/2024]
Abstract
Due to the substantial heterogeneity among extracellular vesicle (EV) subpopulations, single-EV analysis has the potential to elucidate the mechanisms behind EV biogenesis and shed light on the myriad functions, leading to the development of novel diagnostics and therapeutics. While many studies have been devoted to reveal between-EV variations in surface proteins and RNAs, DNA cargos (EV-DNA) have received little attention. Here, we report a hydrogel-based droplet digital multiple displacement amplification approach for the comprehensive analysis of EV-DNA at the single-EV level. Single EVs are dispersed in thousands of hydrogel droplets and lysed for DNA amplification and identification. The droplet microfluidics strategy empowers the assay with single-molecule sensitivity and capability for absolute quantification of DNA-containing EVs. In particular, our findings indicate that 5-40% EVs are associated with DNA, depending on the cell of origin. Large EVs exhibit a higher proportion of DNA-containing EVs and a more substantial presence of intraluminal DNA, compared to small EVs. These DNA-containing EVs carry multiple DNA fragments on average. Furthermore, both double-stranded DNA and single-stranded DNA were able to be detected at the single-EV level. Utilizing this method, the abundance, distribution, and biophysical properties of EV-DNA in various EV populations are evaluated. The DNA level within EVs provides insight into the status of the originating cells and offers valuable information on the outcomes of anticancer treatments. The utilization of single-EV analysis for EV-DNA holds significant promise for early cancer detection and treatment response monitoring.
Collapse
Affiliation(s)
- Yufeng Jiao
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Liyang Gao
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Tao Zhang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| | - Ziyi He
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | | | - Wu Liu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
| |
Collapse
|
30
|
Sun Z, Zhang B, Tu H, Pan C, Chai Y, Chen W. Advances in colorimetric biosensors of exosomes: novel approaches based on natural enzymes and nanozymes. NANOSCALE 2024; 16:1005-1024. [PMID: 38117141 DOI: 10.1039/d3nr05459d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Exosomes are 30-150 nm vesicles derived from diverse cell types, serving as one of the most important biomarkers for early diagnosis and prognosis of diseases. However, the conventional detection method for exosomes faces significant challenges, such as unsatisfactory sensitivity, complicated operation, and the requirement of complicated devices. In recent years, colorimetric exosome biosensors with a visual readout underwent rapid development due to the advances in natural enzyme-based assays and the integration of various types of nanozymes. These synthetic nanomaterials show unique physiochemical properties and catalytic abilities, enabling the construction of exosome colorimetric biosensors with novel principles. This review will illustrate the reaction mechanisms and properties of natural enzymes and nanozymes, followed by a detailed introduction of the recent advances in both types of enzyme-based colorimetric biosensors. A comparison between natural enzymes and nanozymes is made to provide insights into the research that improves the sensitivity and convenience of assays. Finally, the advantages, challenges, and future directions of enzymes as well as exosome colorimetric biosensors are highlighted, aiming at improving the overall performance from different approaches.
Collapse
Affiliation(s)
- Zhonghao Sun
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Binmao Zhang
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Hangjia Tu
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Chuye Pan
- College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518055, China.
| | - Yujuan Chai
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Wenwen Chen
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
31
|
Zhou X, Jia Y, Mao C, Liu S. Small extracellular vesicles: Non-negligible vesicles in tumor progression, diagnosis, and therapy. Cancer Lett 2024; 580:216481. [PMID: 37972701 DOI: 10.1016/j.canlet.2023.216481] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
Small extracellular vesicles (sEVs) such as exosomes are nanoscale membranous particles (<200 nm) that have emerged as crucial targets for liquid biopsy and as promising drug delivery vehicles. They play a significant role in tumor progression as intercellular messengers. They can serve as biomarkers for tumor diagnosis and as drug carriers for cancer treatment. This article reviews recent studies on sEVs in oncology and explores their potential as biomarkers and drug delivery vehicles. Following tumorigenesis, sEVs in the tumor microenvironment (TME) and circulatory system undergo modifications to regulate various events in the TME, including angiogenesis, epithelial-mesenchymal transition (EMT), and tumor immunity, with either pro- or anti-tumor effects. sEVs have been investigated for use as diagnostic and prognostic biomarkers for a variety of tumors, including lung cancer, melanoma, breast cancer, prostate cancer, and hepatocellular carcinoma. sEVs can be used for cancer therapy by packaging drugs or proteins into them through pre- and post-isolation modification techniques. The clinical trials of sEVs as biomarkers and drug carriers are also summarized. Finally, the challenges in the use of sEVs are described and the possible approaches to tackling them are suggested. Overall, sEVs will advance the precision cancer medicine and has shown great potential in clinical applications.
Collapse
Affiliation(s)
- Xinru Zhou
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Yin Jia
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; School of Materials Science & Engineering, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shanrong Liu
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China.
| |
Collapse
|
32
|
Liu C, Xia C, Xia C. Biology and function of exosomes in tumor immunotherapy. Biomed Pharmacother 2023; 169:115853. [PMID: 37951023 DOI: 10.1016/j.biopha.2023.115853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/25/2023] [Accepted: 11/05/2023] [Indexed: 11/13/2023] Open
Abstract
Exosomes are nano-scale extracellular vesicles that are found widely in various biological fluids. As messengers, exosomes deliver characteristic biological information from donor cells, facilitating their accumulation and subsequent transfer of information to tumor immune cells. Immunotherapy is a cutting-edge strategy for cancer therapy, but it has not yet reached its full potential owing to severe side effects and limited efficacy. Exosomes possess antigens and immunostimulatory molecules and can serve as cell-free vaccines to induce antitumor immunity. In addition, given their stability, low immunogenicity, and targeting ability, exosomes represent ideal drug delivery systems in tumor immunotherapy by delivering cargoes, including non-coding ribonucleic acids (RNAs), membrane proteins, chemotherapeutic agents, and immune cell death inducers. Exosomes can also be engineered to precisely target tumor cells. However, as a rising star in tumor immunotherapy, exosomes are also impeded by some challenges, including the lack of uniform technical standards for their isolation and purification, the need to improve exosomal cargo loading for efficient exosome delivery, and the expansion of clinical trials, which are currently in their infancy. Long-term, multi-center, and large-scale clinical trials are needed to evaluate the performance of exosomes in the future. Nonetheless, exosomes have demonstrated encouraging performance in tumor immunotherapy. In this review, we summarize the potential and challenges of exosomes in tumor immunotherapy, with the aim to shed light on exosomes as new-era tumor immunotherapy tools.
Collapse
Affiliation(s)
- Can Liu
- Foshan Maternity and Chlid Healthcare Hospital, Foshan 528000, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 515150, China
| | - Cong Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Chenglai Xia
- Foshan Maternity and Chlid Healthcare Hospital, Foshan 528000, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 515150, China.
| |
Collapse
|
33
|
Liu X, Wang Y, Peng Y, Shi J, Chen W, Wang W, Ma X. Urease-Powered Micromotors with Spatially Selective Distribution of Enzymes for Capturing and Sensing Exosomes. ACS NANO 2023; 17:24343-24354. [PMID: 38038995 DOI: 10.1021/acsnano.3c10405] [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: 12/02/2023]
Abstract
Enzyme-catalyzed micro/nanomotors (MNMs) exhibit tremendous potential for biological isolation and sensing, because of their biocompatibility, versatility, and ready access to biofuel. However, flow field generated by enzyme-catalyzed reactions might significantly hinder performance of surface-linked functional moieties, e.g., the binding interaction between MNMs and target cargos. Herein, we develop enzymatic micromotors with spatially selective distribution of urease to enable the independent operation of various modules and facilitate the capture and sensing of exosomes. When urease is modified into the motors' cavity, the flow field from enzyme catalysis has little effect on the exterior surface of the motors. The active motion and encapsulating urease internally result in enhancement of ∼35% and 18% in binding efficiency of target cargos, e.g., exosomes as an example here, compared to their static counterparts and moving micromotors with urease modified externally, respectively. Once exosomes are trapped, they can be transferred to a clean environment by the motors for Raman signal detection and/or identification using the surface Raman enhancement scattering (SERS) effect of coated gold nanoshell. The biocatalytic micromotors, achieving spatial separation between driving module and function module, offer considerable promise for future design of multifunctional MNMs in biomedicine and diagnostics.
Collapse
Affiliation(s)
- Xiaoxia Liu
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
| | - Yong Wang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yixin Peng
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenjun Chen
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
| | - Wei Wang
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
| | - Xing Ma
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Guangdong, Shenzhen 518055, China
| |
Collapse
|
34
|
Asleh K, Dery V, Taylor C, Davey M, Djeungoue-Petga MA, Ouellette RJ. Extracellular vesicle-based liquid biopsy biomarkers and their application in precision immuno-oncology. Biomark Res 2023; 11:99. [PMID: 37978566 PMCID: PMC10655470 DOI: 10.1186/s40364-023-00540-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
While the field of precision oncology is rapidly expanding and more targeted options are revolutionizing cancer treatment paradigms, therapeutic resistance particularly to immunotherapy remains a pressing challenge. This can be largely attributed to the dynamic tumor-stroma interactions that continuously alter the microenvironment. While to date most advancements have been made through examining the clinical utility of tissue-based biomarkers, their invasive nature and lack of a holistic representation of the evolving disease in a real-time manner could result in suboptimal treatment decisions. Thus, using minimally-invasive approaches to identify biomarkers that predict and monitor treatment response as well as alert to the emergence of recurrences is of a critical need. Currently, research efforts are shifting towards developing liquid biopsy-based biomarkers obtained from patients over the course of disease. Liquid biopsy represents a unique opportunity to monitor intercellular communication within the tumor microenvironment which could occur through the exchange of extracellular vesicles (EVs). EVs are lipid bilayer membrane nanoscale vesicles which transfer a plethora of biomolecules that mediate intercellular crosstalk, shape the tumor microenvironment, and modify drug response. The capture of EVs using innovative approaches, such as microfluidics, magnetic beads, and aptamers, allow their analysis via high throughput multi-omics techniques and facilitate their use for biomarker discovery. Artificial intelligence, using machine and deep learning algorithms, is advancing multi-omics analyses to uncover candidate biomarkers and predictive signatures that are key for translation into clinical trials. With the increasing recognition of the role of EVs in mediating immune evasion and as a valuable biomarker source, these real-time snapshots of cellular communication are promising to become an important tool in the field of precision oncology and spur the recognition of strategies to block resistance to immunotherapy. In this review, we discuss the emerging role of EVs in biomarker research describing current advances in their isolation and analysis techniques as well as their function as mediators in the tumor microenvironment. We also highlight recent lung cancer and melanoma studies that point towards their application as predictive biomarkers for immunotherapy and their potential clinical use in precision immuno-oncology.
Collapse
Affiliation(s)
- Karama Asleh
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada.
| | - Valerie Dery
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
| | - Catherine Taylor
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Michelle Davey
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | | | - Rodney J Ouellette
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- Dr Georges L. Dumont University Hospital, Vitalite Health Network, Moncton, New Brunswick, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| |
Collapse
|
35
|
Chang K, Fang Y, He P, Zhu C, Liu X, Zheng D, Chen D, Liu C. Employing the Anchor DSPE-PEG as a Redox Probe for Ratiometric Electrochemical Detection of Surface Proteins on Extracellular Vesicles with Aptamers. Anal Chem 2023; 95:16194-16200. [PMID: 37889159 DOI: 10.1021/acs.analchem.3c02948] [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: 10/28/2023]
Abstract
Quantitative analysis of surface proteins on extracellular vesicles (EVs) has been considered to be a crucial approach for reflecting the status of diseases. Due to the diverse composition of surface proteins on EVs and the interference from nonvesicular proteins, accurately detecting the expression of surface proteins on EVs remains a challenging task. While membrane affinity molecules have been widely employed as EVs capture probes to address this issue, their inherent biochemical properties have not been effectively harnessed. In this paper, we found that the electrochemical redox activity of the DSPE-PEG molecule was diminished upon its insertion into the membrane of EVs. This observation establishes the DSPE-PEG molecule modified on the Au electrode surface as a capture and a redox probe for the electrochemical detection of EVs. By utilizing methylene blue-labeled aptamers, the targeted surface proteins of EVs can be detected by recording the ratio of the oxidation peak current of methylene blue and DSPE-PEG. Without complicated signal amplification, the detection limit for EVs is calculated to be 8.11 × 102 particles/mL. Using this platform, we directly analyzed the expression of CD63 and HER2 proteins on the surface of EVs in human clinical plasma samples, demonstrating its significant potential in distinguishing breast cancer patients from healthy individuals.
Collapse
Affiliation(s)
- Kaili Chang
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Yi Fang
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Ping He
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Chunnan Zhu
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Xiaojun Liu
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Dongyun Zheng
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Dongjuan Chen
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| | - Chao Liu
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| |
Collapse
|
36
|
Zhang Q, Chen M, Xu F, Wu W, Luo X, Wang Y, Li J, Cui X, Tan Y, Li Z, Lin Y, Zhang H, Wang W. One-pot preparation of bi-functional POSS-based hybrid monolith via photo-initiated polymerization for isolation of extracellular vesicles. Anal Chim Acta 2023; 1279:341785. [PMID: 37827681 DOI: 10.1016/j.aca.2023.341785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/16/2023] [Accepted: 09/05/2023] [Indexed: 10/14/2023]
Abstract
Extracellular vesicles (EVs) are important participants in numerous pathophysiological processes, and could be used as valuable biomarkers to detect and monitor various diseases. However, facile EV isolation methods are the essential and preliminary issue for their downstream analysis and function investigation. In this work, a polyhedral oligomeric silsesquioxanes (POSS) based hybrid monolith combined metal affinity chromatography (MAC) and distearoyl phospholipid ethanolamine (DSPE) function was developed via photo-initiated thiol-ene polymerization. This synthesis process was facile, simple and convenient, and the obtained hybrid monolith could be applied to efficiently isolate EVs from bio-samples by taking advantages of the specific bond of Ti4+ and phosphate groups on the phospholipid membrane of EVs and the synergistic effect of DSPE insertion. Meanwhile, the eluted EVs could maintain their structural integrity and biological activity, suggesting they could be used for downstream application. Furthermore, 75 up-regulated proteins and 56 down-regulated proteins were identified by comparing the urinary EVs of colorectal cancer (CRC) patients and healthy donors, and these proteins might be used as potential biomarkers for early screening of CRC. These results demonstrated that this hybrid monolith could be used as a simple and convenient tool for isolating EVs from bio-samples and for wider applications in biomarker discovery.
Collapse
Affiliation(s)
- Qi Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Fang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Wen Wu
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xintong Luo
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ying Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China; Taichang Liuhe People's Hospital, Suzhou, 215431, China
| | - Jiaxi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Xuanhao Cui
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yujia Tan
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yujie Lin
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Haiyang Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
37
|
Chen K, Duong BTV, Ahmed SU, Dhavarasa P, Wang Z, Labib M, Flynn C, Xu J, Zhang YY, Wang H, Yang X, Das J, Zargartalebi H, Ma Y, Kelley SO. A magneto-activated nanoscale cytometry platform for molecular profiling of small extracellular vesicles. Nat Commun 2023; 14:5576. [PMID: 37696888 PMCID: PMC10495366 DOI: 10.1038/s41467-023-41285-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
Exosomal PD-L1 (exoPD-L1) has recently received significant attention as a biomarker predicting immunotherapeutic responses involving the PD1/PD-L1 pathway. However, current technologies for exosomal analysis rely primarily on bulk measurements that do not consider the heterogeneity found within exosomal subpopulations. Here, we present a nanoscale cytometry platform NanoEPIC, enabling phenotypic sorting and exoPD-L1 profiling from blood plasma. We highlight the efficacy of NanoEPIC in monitoring anti-PD-1 immunotherapy through the interrogation of exoPD-L1. NanoEPIC generates signature exoPD-L1 patterns in responders and non-responders. In mice treated with PD1-targeted immunotherapy, exoPD-L1 is correlated with tumor growth, PD-L1 burden in tumors, and the immune suppression of CD8+ tumor-infiltrating lymphocytes. Small extracellular vesicles (sEVs) with different PD-L1 expression levels display distinctive inhibitory effects on CD8 + T cells. NanoEPIC offers robust, high-throughput profiling of exosomal markers, enabling sEV subpopulation analysis. This platform holds the potential for enhanced cancer screening, personalized treatment, and therapeutic response monitoring.
Collapse
Affiliation(s)
- Kangfu Chen
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Bill T V Duong
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Sharif U Ahmed
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | | | - Zongjie Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, Northwestern University, Evanston, IL, USA
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth, UK
| | - Connor Flynn
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Jingya Xu
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - Yi Y Zhang
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Hansen Wang
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Xiaolong Yang
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Jagotamoy Das
- Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Hossein Zargartalebi
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Yuan Ma
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Shana O Kelley
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.
- Department of Chemistry, University of Toronto, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA.
| |
Collapse
|
38
|
He J, Li H, Mai J, Ke Y, Zhai C, Li JJ, Jiang L, Shen G, Ding X. Profiling extracellular vesicle surface proteins with 10 µL peripheral plasma within 4 h. J Extracell Vesicles 2023; 12:e12364. [PMID: 37654045 PMCID: PMC10471920 DOI: 10.1002/jev2.12364] [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: 03/01/2023] [Revised: 07/19/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
Extracellular vesicle (EV) surface proteins, expressed by primary tumours, are important biomarkers for early cancer diagnosis. However, the detection of these EV proteins is complicated by their low abundance and interference from non-EV components in clinical samples. Herein, we present a MEmbrane-Specific Separation and two-step Cascade AmpLificatioN (MESS2CAN) strategy for direct detection of EV surface proteins within 4 h. MESS2CAN utilises novel lipid probes (long chains linked by PEG2K with biotin at one end, and DSPE at the other end) and streptavidin-coated magnetic beads, permitting a 49.6% EV recovery rate within 1 h. A dual amplification strategy with a primer exchange reaction (PER) cascaded by the Cas12a system then allows sensitive detection of the target protein at 10 EV particles per microliter. Using 4 cell lines and 90 clinical test samples, we demonstrate MESS2CAN for analysing HER2, EpCAM and EGFR expression on EVs derived from cells and patient plasma. MESS2CAN reports the desired specificity and sensitivity of EGFR (AUC = 0.98) and of HER2 (AUC = 1) for discriminating between HER2-positive breast cancer, triple-negative breast cancer and healthy donors. MESS2CAN is a pioneering method for highly sensitive in vitro EV diagnostics, applicable to clinical samples with trace amounts of EVs.
Collapse
Affiliation(s)
- Jie He
- Department of Anesthesiology and Surgical Intensive Care Unit School of Medicine and School of Biomedical EngineeringXinhua Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Hengyu Li
- Department of Breast and Thyroid SurgeryChanghai Hospital, Naval Military Medical UniversityShanghaiChina
| | - John Mai
- Alfred E. Mann Institute for Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Yuqing Ke
- Department of Anesthesiology and Surgical Intensive Care Unit School of Medicine and School of Biomedical EngineeringXinhua Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Chunhui Zhai
- Department of Anesthesiology and Surgical Intensive Care Unit School of Medicine and School of Biomedical EngineeringXinhua Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jiao Jiao Li
- School of Biomedical Engineering Faculty of Engineering and ITUniversity of Technology SydneySydneyNSWAustralia
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit School of Medicine and School of Biomedical EngineeringXinhua Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Guangxia Shen
- Department of Anesthesiology and Surgical Intensive Care Unit School of Medicine and School of Biomedical EngineeringXinhua Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit School of Medicine and School of Biomedical EngineeringXinhua Hospital, Shanghai Jiao Tong UniversityShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
39
|
Ma H, Xing F, Zhou Y, Yu P, Luo R, Xu J, Xiang Z, Rommens PM, Duan X, Ritz U. Design and fabrication of intracellular therapeutic cargo delivery systems based on nanomaterials: current status and future perspectives. J Mater Chem B 2023; 11:7873-7912. [PMID: 37551112 DOI: 10.1039/d3tb01008b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Intracellular cargo delivery, the introduction of small molecules, proteins, and nucleic acids into a specific targeted site in a biological system, is an important strategy for deciphering cell function, directing cell fate, and reprogramming cell behavior. With the advancement of nanotechnology, many researchers use nanoparticles (NPs) to break through biological barriers to achieving efficient targeted delivery in biological systems, bringing a new way to realize efficient targeted drug delivery in biological systems. With a similar size to many biomolecules, NPs possess excellent physical and chemical properties and a certain targeting ability after functional modification on the surface of NPs. Currently, intracellular cargo delivery based on NPs has emerged as an important strategy for genome editing regimens and cell therapy. Although researchers can successfully deliver NPs into biological systems, many of them are delivered very inefficiently and are not specifically targeted. Hence, the development of efficient, target-capable, and safe nanoscale drug delivery systems to deliver therapeutic substances to cells or organs is a major challenge today. In this review, on the basis of describing the research overview and classification of NPs, we focused on the current research status of intracellular cargo delivery based on NPs in biological systems, and discuss the current problems and challenges in the delivery process of NPs in biological systems.
Collapse
Affiliation(s)
- Hong Ma
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Fei Xing
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Ludwigstraße 23, 35392 Giessen, Germany
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Rong Luo
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiawei Xu
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Xin Duan
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedic Surgery, The Fifth People's Hospital of Sichuan Province, Chengdu, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
40
|
Pan S, Ding A, Li Y, Sun Y, Zhan Y, Ye Z, Song N, Peng B, Li L, Huang W, Shao H. Small-molecule probes from bench to bedside: advancing molecular analysis of drug-target interactions toward precision medicine. Chem Soc Rev 2023; 52:5706-5743. [PMID: 37525607 DOI: 10.1039/d3cs00056g] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Over the past decade, remarkable advances have been witnessed in the development of small-molecule probes. These molecular tools have been widely applied for interrogating proteins, pathways and drug-target interactions in preclinical research. While novel structures and designs are commonly explored in probe development, the clinical translation of small-molecule probes remains limited, primarily due to safety and regulatory considerations. Recent synergistic developments - interfacing novel chemical probes with complementary analytical technologies - have introduced and expedited diverse biomedical opportunities to molecularly characterize targeted drug interactions directly in the human body or through accessible clinical specimens (e.g., blood and ascites fluid). These integrated developments thus offer unprecedented opportunities for drug development, disease diagnostics and treatment monitoring. In this review, we discuss recent advances in the structure and design of small-molecule probes with novel functionalities and the integrated development with imaging, proteomics and other emerging technologies. We further highlight recent applications of integrated small-molecule technologies for the molecular analysis of drug-target interactions, including translational applications and emerging opportunities for whole-body imaging, tissue-based measurement and blood-based analysis.
Collapse
Affiliation(s)
- Sijun Pan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yisi Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yaxin Sun
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Yueqin Zhan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Zhenkun Ye
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Ning Song
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
| | - Wei Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China.
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore 117599, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore
| |
Collapse
|
41
|
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
|
42
|
Nguyen CM, Sallam M, Islam MS, Clack K, Soda N, Nguyen NT, Shiddiky MJA. Placental Exosomes as Biomarkers for Maternal Diseases: Current Advances in Isolation, Characterization, and Detection. ACS Sens 2023. [PMID: 37449399 DOI: 10.1021/acssensors.3c00689] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Serving as the interface between fetal and maternal circulation, the placenta plays a critical role in fetal growth and development. Placental exosomes are small membrane-bound extracellular vesicles released by the placenta during pregnancy. They contain a variety of biomolecules, including lipids, proteins, and nucleic acids, which can potentially be biomarkers of maternal diseases. An increasing number of studies have demonstrated the utility of placental exosomes for the diagnosis and monitoring of pathological conditions such as pre-eclampsia and gestational diabetes. This suggests that placental exosomes may serve as new biomarkers in liquid biopsy analysis. This review provides an overview of the current understanding of the biological function of placental exosomes and their potential as biomarkers of maternal diseases. Additionally, this review highlights current barriers and the way forward for standardization and validation of known techniques for exosome isolation, characterization, and detection. Finally, microfluidic devices for exosome research are discussed.
Collapse
Affiliation(s)
- Cong Minh Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Mohamed Sallam
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Md Sajedul Islam
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Southport, QLD 4222, Australia
| | - Kimberley Clack
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Narshone Soda
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Muhammad J A Shiddiky
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Rural Health Research Institute, Charles Sturt University, Orange, NSW 2800, Australia
| |
Collapse
|
43
|
Zheng J, Hu X, Zeng Y, Zhang B, Sun Z, Liu X, Zheng W, Chai Y. Review of the advances in lipid anchors-based biosensors for the isolation and detection of exosomes. Anal Chim Acta 2023; 1263:341319. [PMID: 37225343 DOI: 10.1016/j.aca.2023.341319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/26/2023]
Abstract
Exosomes are nanoparticles with a bilayer lipid structure that carry cargo from their cells of origin. These vesicles are vital to disease diagnosis and therapeutics; however, conventional isolation and detection techniques are generally complicated, time-consuming, and costly, thus hampering the clinical applications of exosomes. Meanwhile, sandwich-structured immunoassays for exosome isolation and detection rely on the specific binding of membrane surface biomarkers, which may be limited by the type and amount of target protein present. Recently, lipid anchors inserted into the membranes of vesicles through hydrophobic interactions have been adopted as a new strategy for extracellular vesicle manipulation. By combining nonspecific and specific binding, the performance of biosensors can be improved variously. This review presents the reaction mechanisms and properties of lipid anchors/probes, as well as advances in the development of biosensors. The combination of signal amplification methods with lipid anchors is discussed in detail to provide insights into the design of convenient and sensitive detection techniques. Finally, the advantages, challenges, and future directions of lipid anchor-based exosome isolation and detection methods are highlighted from the perspectives of research, clinical use, and commercialization.
Collapse
Affiliation(s)
- Junyuan Zheng
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Xiaoxiang Hu
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Yuping Zeng
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Binmao Zhang
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Zhonghao Sun
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Xiaowei Liu
- Department of Management, Shenzhen University, Shenzhen, 518055, China.
| | - Weidong Zheng
- Department of Laboratory Medicine, Shenzhen University General Hospital, Shenzhen, 518055, China.
| | - Yujuan Chai
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
44
|
Cui M, Dutcher S, Bayly P, Meacham J. Robust acoustic trapping and perturbation of single-cell microswimmers illuminate three-dimensional swimming and ciliary coordination. Proc Natl Acad Sci U S A 2023; 120:e2218951120. [PMID: 37307440 PMCID: PMC10290211 DOI: 10.1073/pnas.2218951120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 04/18/2023] [Indexed: 06/14/2023] Open
Abstract
We report a label-free acoustic microfluidic method to confine single, cilia-driven swimming cells in space without limiting their rotational degrees of freedom. Our platform integrates a surface acoustic wave (SAW) actuator and bulk acoustic wave (BAW) trapping array to enable multiplexed analysis with high spatial resolution and trapping forces that are strong enough to hold individual microswimmers. The hybrid BAW/SAW acoustic tweezers employ high-efficiency mode conversion to achieve submicron image resolution while compensating for parasitic system losses to immersion oil in contact with the microfluidic chip. We use the platform to quantify cilia and cell body motion for wildtype biciliate cells, investigating effects of environmental variables like temperature and viscosity on ciliary beating, synchronization, and three-dimensional helical swimming. We confirm and expand upon the existing understanding of these phenomena, for example determining that increasing viscosity promotes asynchronous beating. Motile cilia are subcellular organelles that propel microorganisms or direct fluid and particulate flow. Thus, cilia are critical to cell survival and human health. The unicellular alga Chlamydomonas reinhardtii is widely used to investigate the mechanisms underlying ciliary beating and coordination. However, freely swimming cells are difficult to image with sufficient resolution to capture cilia motion, necessitating that the cell body be held during experiments. Acoustic confinement is a compelling alternative to use of a micropipette, or to magnetic, electrical, and optical trapping that may modify the cells and affect their behavior. Beyond establishing our approach to studying microswimmers, we demonstrate a unique ability to mechanically perturb cells via rapid acoustic positioning.
Collapse
Affiliation(s)
- Mingyang Cui
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Susan K. Dutcher
- Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO63110
| | - Philip V. Bayly
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
| | - J. Mark Meacham
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO63130
| |
Collapse
|
45
|
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
|
46
|
Deng D, Li X, Zhang JJ, Yin Y, Tian Y, Gan D, Wu R, Wang J, Tian BM, Chen FM, He XT. Biotin-Avidin System-Based Delivery Enhances the Therapeutic Performance of MSC-Derived Exosomes. ACS NANO 2023; 17:8530-8550. [PMID: 37115712 DOI: 10.1021/acsnano.3c00839] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Exosomes (EXs) shed by mesenchymal stem cells (MSCs) are potent therapeutic agents that promote wound healing and regeneration, but when used alone in vivo, their therapeutic potency is diminished by rapid clearance and bioactivity loss. Inspired by the biotin-avidin interaction, we developed a simple yet versatile method for the immobilization of MSC-derived EXs (MSC-EXs) into hydrogels and achieved sustained release for regenerative purposes. First, biotin-modified gelatin methacryloyl (Bio-GelMA) was fabricated by grafting NHS-PEG12-biotin onto the amino groups of GelMA. Biotin-modified MSC-EXs (Bio-EXs) were then synthesized using an in situ self-assembling biotinylation strategy, which provided sufficient binding sites for MSC-EX delivery with little effect on their cargo composition. Thereafter, Bio-EXs were immobilized in Bio-GelMA via streptavidin to generate Bio-GelMA@Bio-EX hydrogels. An in vitro analysis demonstrated that Bio-EXs could be taken up by macrophages and exerted immunomodulatory effects similar to those of MSC-EXs, and Bio-GelMA@Bio-EX hydrogels provided sustained release of MSC-EXs for 7 days. After subcutaneous transplantation, a more constant retention of MSC-EXs in Bio-GelMA@Bio-EX hydrogels was observed for up to 28 days. When placed in an artificial periodontal multitissue defect, the functionalized hydrogels exhibited an optimized therapeutic performance to regrow complex periodontal tissues, including acellular cementum, periodontal ligaments (PDLs), and alveolar bone. In this context, Bio-GelMA@Bio-EX hydrogels exerted a robust immunomodulatory effect that promoted macrophage polarization toward an M2 phenotype. Our findings demonstrate that MSC-EXs delivered with the aid of the biotin-avidin system exhibit robust macrophage-modulating and repair-promoting functions and suggest a universal approach for the development of MSC-EX-functionalized biomaterials for advanced therapies.
Collapse
Affiliation(s)
- Daokun Deng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Xuan Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Jiu-Jiu Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Yi Tian
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Dian Gan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Ruixin Wu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Jia Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Bei-Min Tian
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Xiao-Tao He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| |
Collapse
|
47
|
Gajula SNR, Nalla LV. Small extracellular vesicle biomarkers in breast cancer: a real-time snapshot for early diagnosis and prognosis? Bioanalysis 2023; 15:367-370. [PMID: 37154153 DOI: 10.4155/bio-2023-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Affiliation(s)
- Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research, Hyderabad, Telangana, 500037, India
| | - Lakshmi Vineela Nalla
- Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, 522302, India
| |
Collapse
|
48
|
Li Q, Zhang Z, Wang F, Wang X, Zhan S, Yang X, Xu C, Liu D. Reversible zwitterionic coordination enables rapid, high-yield, and high-purity isolation of extracellular vesicles from biofluids. SCIENCE ADVANCES 2023; 9:eadf4568. [PMID: 37058564 PMCID: PMC10104463 DOI: 10.1126/sciadv.adf4568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 03/10/2023] [Indexed: 06/19/2023]
Abstract
Extracellular vesicles (EVs) hold great clinical value as promising diagnostic biomarkers and therapeutic agents. This field, however, is hindered by technical challenges in the isolation of EVs from biofluids for downstream purposes. We here report a rapid (<30 min) isolation method for EV extraction from diverse biofluids with yield and purity exceeding 90%. These high performances are ascribed to the reversible zwitterionic coordination between the phosphatidylcholine (PC) on EV membranes and the "PC-inverse" choline phosphate (CP) decorated on magnetic beads. By coupling this isolation method with proteomics, a set of differentially expressed proteins on the EVs were identified as potential colon cancer biomarkers. Last, we demonstrated that the EVs in various clinically relevant biofluids, such as blood serum, urine, and saliva, can also be isolated efficiently, outperforming the conventional approaches in terms of simplicity, speed, yield, and purity.
Collapse
Affiliation(s)
- Qiang Li
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Sciences, Tianjin Key Laboratory of Molecular Recognition and Biosensing, and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhaowei Zhang
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Sciences, Tianjin Key Laboratory of Molecular Recognition and Biosensing, and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Fengchao Wang
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Sciences, Tianjin Key Laboratory of Molecular Recognition and Biosensing, and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiang Wang
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Sciences, Tianjin Key Laboratory of Molecular Recognition and Biosensing, and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Saisong Zhan
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Sciences, Tianjin Key Laboratory of Molecular Recognition and Biosensing, and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaoqing Yang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Chen Xu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin Institute of Coloproctology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Dingbin Liu
- State Key Laboratory of Medicinal Chemical Biology, Research Center for Analytical Sciences, Tianjin Key Laboratory of Molecular Recognition and Biosensing, and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
49
|
Parvin R, Zhang L, Zu Y, Ye F. Photothermal Responsive Digital Polymerase Chain Reaction Resolving Exosomal microRNAs Expression in Liver Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2207672. [PMID: 36942691 DOI: 10.1002/smll.202207672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/05/2023] [Indexed: 06/18/2023]
Abstract
Exosomal microRNAs have been studied as a good source of noninvasive biomarkers due to their functions in genetic exchange between cells and have been already well documented in many biological activities; however, inaccuracy remains a key challenge for liver cancer surveillance. Herein, a versatile duplex photothermal digital polymerase chain reaction (PCR) strategy combined with a lipid nanoparticle-based exosome capture approach is proposed to profile microRNAs expression through a 3-h easy-to-operate process. The microfluidically-generated molybdenum disulfide-nanocomposite-doped gelatin microcarriers display attractive properties as a 2-4 °C s-1 ramping-up rate triggered by near-infrared and reversible sol-gel transforming in step with PCR activation. To achieve PCR thermocycling, the corresponding irradiation coordinating with fan cooling are automatically performed via a homemade control module with programs. Thus, taking the multiplexing capability of dual-color labeling, 19-31 folds higher in exosomal microRNA-200b-3p and microRNA-21-5p, and tenfold lower in microRNA-22-3p expressions relative to the control microRNA-26a-5p are quantified in two liver cancer cells (Huh7 and HepG2) than in those from the healthy cells. It is believed that this exosomal microRNA genotyping method would be highly applicable for liver cancer diagnostics.
Collapse
Affiliation(s)
- Rokshana Parvin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
| | - Lexiang Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
| | - Yan Zu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
| | - Fangfu Ye
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| |
Collapse
|
50
|
Xia H, Yu Z, Zhang L, Liu S, Zhao Y, Huang J, Fu D, Xie Q, Liu H, Zhang Z, Zhao Y, Wu M, Zhang W, Pang D, Chen G. Real-Time Dissection of the Transportation and miRNA-Release Dynamics of Small Extracellular Vesicles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205566. [PMID: 36599707 PMCID: PMC9982592 DOI: 10.1002/advs.202205566] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/28/2022] [Indexed: 06/17/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-enclosed structures that deliver biomolecules for intercellular communication. Developing visualization methods to elucidate the spatiotemporal dynamics of EVs' behaviors will facilitate their understanding and translation. With a quantum dot (QD) labeling strategy, a single particle tracking (SPT) platform is proposed here for dissecting the dynamic behaviors of EVs. The interplays between tumor cell-derived small EVs (T-sEVs) and endothelial cells (ECs) are specifically investigated based on this platform. It is revealed that, following a clathrin-mediated endocytosis by ECs, T-sEVs are transported to the perinuclear region in a typical three-stage pattern. Importantly, T-sEVs frequently interact with and finally enter lysosomes, followed by quick release of their carried miRNAs. This study, for the first time, reports the entire process and detailed dynamics of T-sEV transportation and cargo-release in ECs, leading to better understanding of their proangiogenic functions. Additionally, the QD-based SPT technique will help uncover more secrets of sEV-mediated cell-cell communication.
Collapse
Affiliation(s)
- Hou‐Fu Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
- Department of Oral and Maxillofacial SurgerySchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Zi‐Li Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
- Department of Oral and Maxillofacial SurgerySchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Li‐Juan Zhang
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072P. R. China
| | - Shu‐Lin Liu
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Biosensing and Molecular RecognitionResearch Center for Analytical Sciencesand College of ChemistryNankai UniversityTianjin300071P. R. China
| | - Yi Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
- Department of ProsthodonticsSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Jue Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Dan‐Dan Fu
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072P. R. China
| | - Qi‐Hui Xie
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Hai‐Ming Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Zhi‐Ling Zhang
- College of Chemistry and Molecular SciencesWuhan UniversityWuhan430072P. R. China
| | - Yi‐Fang Zhao
- Department of Oral and Maxillofacial SurgerySchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Min Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Wei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
- Department of Oral and Maxillofacial SurgerySchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
| | - Dai‐Wen Pang
- State Key Laboratory of Medicinal Chemical BiologyTianjin Key Laboratory of Biosensing and Molecular RecognitionResearch Center for Analytical Sciencesand College of ChemistryNankai UniversityTianjin300071P. R. China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral Biomedicine Ministry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
- Department of Oral and Maxillofacial SurgerySchool and Hospital of StomatologyWuhan UniversityWuhan430079P. R. China
- TaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430071P. R. China
- Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhan430071P. R. China
| |
Collapse
|