1
|
Stanley CV, Xiao Y, Ling T, Li DS, Chen P. Opto-digital molecular analytics. Chem Soc Rev 2025; 54:3557-3577. [PMID: 40035639 DOI: 10.1039/d5cs00023h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
In contrast to conventional ensemble-average-based methods, opto-digital molecular analytic approaches digitize detection by physically partitioning individual detection events into discrete compartments or directly locating and analyzing the signals from single molecules. The sensitivity can be enhanced by signal amplification reactions, signal enhancement interactions, labelling by strong signal emitters, advanced optics, image processing, and machine learning, while specificity can be improved by designing target-selective probes and profiling molecular dynamics. With the capabilities to attain a limit of detection several orders lower than the conventional methods, reveal intrinsic molecular information, and achieve multiplexed analysis using a small-volume sample, the emerging opto-digital molecular analytics may be revolutionarily instrumental to clinical diagnosis, molecular chemistry and science, drug discovery, and environment monitoring. In this article, we provide a comprehensive review of the recent advances, offer insights into the underlying mechanisms, give comparative discussions on different strategies, and discuss the current challenges and future possibilities.
Collapse
Affiliation(s)
- Chelsea Violita Stanley
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore.
- Lee Kong Chian School of Medicine, Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| | - Yi Xiao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore.
- Lee Kong Chian School of Medicine, Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| | - Tong Ling
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore.
| | - Dong-Sheng Li
- College of Materials and Chemical Engineering, Key Laboratory of Inorganic Nonmetallic Crystalline and Energy Conversion Materials, China Three Gorges University, Yichang, 443002, P. R. China
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore.
- Lee Kong Chian School of Medicine, Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
- Skin Research Institute of Singapore, 308232, Singapore
| |
Collapse
|
2
|
Liu J, Srivastava S, Li T, Moujane F, Lee JY, Chen Y, Liu H, Deng SX, Xie YH. On the Feasibility of SERS-Based Monitoring of Drug Loading Efficiency in Exosomes for Targeted Delivery. BIOSENSORS 2025; 15:141. [PMID: 40136938 PMCID: PMC11939968 DOI: 10.3390/bios15030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025]
Abstract
Cancer, a significant cause of mortality, necessitates improved drug delivery strategies. Exosomes, as natural drug carriers, offer a more efficient, targeted, and less toxic drug delivery system compared to direct dispersal methods via ingestion or injection. To be successfully implemented as drug carriers, efficient loading of drugs into exosomes is crucial, and a deeper understanding of the loading mechanism remains to be solved. This study introduces surface-enhanced Raman scattering (SERS) to monitor drug loading efficacy at the single vesicle level. By enhancing the Raman signal, SERS overcomes limitations in Raman spectroscopy. A gold nanopyramids array-based SERS substrate assesses exosome heterogeneity in drug-loading capabilities with the help of single-layer graphene for precise quantification. This research advances targeted drug delivery by presenting a more efficient method of evaluating drug-loading efficiency into individual exosomes through SERS-based monitoring. Furthermore, the study explores leveraging osmotic pressure variations, enhancing the efficiency of drug loading into exosomes.
Collapse
Affiliation(s)
- Jun Liu
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Siddharth Srivastava
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tieyi Li
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Faycal Moujane
- Cornea Division, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - John Y. Lee
- Cornea Division, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yiqing Chen
- Department of Bioengineering, University of California Riverside, Riverside, CA 92521, USA
| | - Huinan Liu
- Department of Bioengineering, University of California Riverside, Riverside, CA 92521, USA
| | - Sophie X. Deng
- Cornea Division, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ya-Hong Xie
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Chen B, Qiu X. Surface-Enhanced Raman Scattering (SERS) for exosome detection. Clin Chim Acta 2025; 568:120148. [PMID: 39842651 DOI: 10.1016/j.cca.2025.120148] [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: 12/16/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Exosomes, nanoscale extracellular vesicles secreted by various cells, are abundantly present in biological fluids. They have been identified as carriers of specific molecules, suggesting their potential role in early disease detection. However, their clinical application is hindered by several challenges, including the need for large sample volumes for enrichment, limitations of traditional detection methods, and the complexity involved in phenotype analysis and separation. OBJECTIVE This review aims to explore the application of Surface-Enhanced Raman Scattering (SERS) technology in exosome detection. SERS, known for its unique photonic properties and high sensitivity, offers a promising solution for detecting exosomes without the need for large sample volumes or extensive phenotypic analysis. This review focuses on the real-time and non-invasive assessment capabilities of SERS in exosome detection, providing insights into its potential for early disease diagnosis. CONCLUSION The review concludes by emphasizing the potential of SERS-based exosome detection in advancing early disease diagnosis. By overcoming existing challenges, SERS technology offers a promising approach for the development of sensitive and specific diagnostic assays, contributing to better patient outcomes and personalized medicine.
Collapse
Affiliation(s)
- Biqing Chen
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Heilongjiang 150081 PR China
| | - Xiaohong Qiu
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Heilongjiang 150081 PR China.
| |
Collapse
|
4
|
Li A, Mo X, Lu Y, Zhu G, Liu C, Yang X, Huang Y, Sheng J, Zhang H, Meng D, Zhao X. Digital SERS immunoassay of Interleukin-6 based on Au@Ag-Au nanotags. Biosens Bioelectron 2025; 270:116973. [PMID: 39581067 DOI: 10.1016/j.bios.2024.116973] [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: 09/07/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Interleukin-6 (IL-6) is a crucial cytokine involved in inflammation and immune regulation. However, the detection of IL-6 with ultrasensitivity and high specificity remains a significant challenge due to the inherent complexity of biofluids. Herein, we present a digital surface enhanced Raman scattering (SERS) immunoassay using core-shell Au@Ag-Au nanotags for IL-6 detection with ultrasensitivity and high reliability. A low-cost silicon chip was functionalized as capture substrates, employing novel SERS nanotags that exhibit strong, robust and reproducible signals at single-nanoparticle resolution as the amplification element. We proposed two analytical methods to validate single-molecule events follow a Poisson distribution and to quantify protein biomarkers over a broad linear dynamic range, respectively. The strong alignment between theoretical and experimental results enhances the method's reliability. Our assay provides two readouts: colorimetric analysis by naked eyes for high concentrations (>1 ng/mL) and digital SERS analysis for low concentrations. Following method optimization, we obtained a linear range from 100 fg/mL to 1 ng/mL (R2 = 0.994) with a limit of detection (LOD) of 12.4 fg/mL, suitable for clinical applications. The method was tested for IL-6 quantification in healthy human serum and saliva, with recoveries from 92.4% to 105.3%. Finally, the immunoassay demonstrated strong consistency with the standard clinical laboratory method when tested with clinical serum samples. Thus, our proposed the digital SERS immunoassay is a promising tool for the precision clinical diagnosis of IL-6-related diseases or other conditions.
Collapse
Affiliation(s)
- Ao Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China
| | - Xiufang Mo
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China
| | - Yu Lu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China
| | - Geng Zhu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China
| | - Chang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China
| | - Xi Yang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China
| | - Yan Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China
| | - Jinliang Sheng
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Hui Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, 832003, China
| | - Dianhuai Meng
- Rehabilitation Medical Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Xiangwei Zhao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China; Southeast University Shenzhen Research Institute, Shenzhen, 518000, China; Institute of Biomaterials and Medical Devices, Southeast University, Suzhou, 215163, China.
| |
Collapse
|
5
|
Lin X, Zhu J, Shen J, Zhang Y, Zhu J. Advances in exosome plasmonic sensing: Device integration strategies and AI-aided diagnosis. Biosens Bioelectron 2024; 266:116718. [PMID: 39216205 DOI: 10.1016/j.bios.2024.116718] [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: 06/14/2024] [Revised: 08/11/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Exosomes, as next-generation biomarkers, has great potential in tracking cancer progression. They face many detection limitations in cancer diagnosis. Plasmonic biosensors have attracted considerable attention at the forefront of exosome detection, due to their label-free, real-time, and high-sensitivity features. Their advantages in multiplex immunoassays of minimal liquid samples establish the leading position in various diagnostic studies. This review delineates the application principles of plasmonic sensing technologies, highlighting the importance of exosomes-based spectrum and image signals in disease diagnostics. It also introduces advancements in miniaturizing plasmonic biosensing platforms of exosomes, which can facilitate point-of-care testing for future healthcare. Nowadays, inspired by the surge of artificial intelligence (AI) for science and technology, more and more AI algorithms are being adopted to process the exosome spectrum and image data from plasmonic detection. Using representative algorithms of machine learning has become a mainstream trend in plasmonic biosensing research for exosome liquid biopsy. Typically, these algorithms process complex exosome datasets efficiently and establish powerful predictive models for precise diagnosis. This review further discusses critical strategies of AI algorithm selection in exosome-based diagnosis. Particularly, we categorize the AI algorithms into the interpretable and uninterpretable groups for exosome plasmonic detection applications. The interpretable AI enhances the transparency and reliability of diagnosis by elucidating the decision-making process, while the uninterpretable AI provides high diagnostic accuracy with robust data processing by a "black-box" working mode. We believe that AI will continue to promote significant progress of exosome plasmonic detection and mobile healthcare in the near future.
Collapse
Affiliation(s)
- Xiangyujie Lin
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaheng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaqing Shen
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Youyu Zhang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
6
|
Yang K, Fu W, Ma Y, Wu M, Li X, Wang Y. A simple capillary isoelectric focusing method as the novel strategy for the isoelectric point measurement of exosomes and its application in disease diagnosis. Mikrochim Acta 2024; 192:8. [PMID: 39636339 DOI: 10.1007/s00604-024-06864-6] [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: 09/18/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
A straightforward capillary isoelectric focusing (cIEF) method is established with a isoelectric point (pI) range spanning from 3.5 to 7.0, exhibiting excellent linearity and repeatability, with an R2 value of 0.9937 and migration time RSDs for all standard proteins below 0.3%. Subsequently, this method was applied to model exosomes derived from cell lines and healthy human serum, and the peak attributions of exosomes were identified using DiI labeled exosomes and lysed exosomes. The reproducibility of this method in exosome detection was also validated, as the RSDs of all pI values were less than 1%. Then, we observed a significant increase in the pI of exosomes with higher cholesterol content, irrespective of whether they originated from cell culture medium or mouse plasma. Notably, serum exosomes from healthy volunteers exhibited higher pI values compared to those from hepatocellular carcinoma patients, suggesting a potential diagnostic perspective for cancer. These findings underscore the significance of pI measurement in reflecting modifications in exosomal lipid membrane composition and their implications in biological functions mediated by exosomes.
Collapse
Affiliation(s)
- Kaige Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wenchang Fu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yinjie Ma
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mingyuan Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinyan Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
7
|
Park JE, Nam H, Hwang JS, Kim S, Kim SJ, Kim S, Jeon JS, Yang M. Label-Free Exosome Analysis by Surface-Enhanced Raman Scattering Spectroscopy with Laser-Ablated Silver Nanoparticle Substrate. Adv Healthc Mater 2024; 13:e2402038. [PMID: 39318105 DOI: 10.1002/adhm.202402038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Early diagnostics of breast cancer is crucial to reduce the risk of cancer metastasis and late relapse. Exosome, which contains distinct information of its origin, can be the target object as a liquid biopsy. However, its low sensitivity and inadequate diagnostic tools interfere with the point-of-care testing (POCT) of the exosome. Recently, Surface-enhanced Raman Scattering (SERS) spectroscopy, which amplifies the Raman scattering, has been proved as a promising tool for exosome detection. However, the fabrication process of SERS probe or substrate is still inefficient and far from large-scale production. This study proposes rapid and label-free detection of breast cancer-derived exosomes by statistical analysis of SERS spectra using silver-nanoparticle-based SERS substrate fabricated by selective laser ablation and melting (SLAM). Employing silver nanowires and optimizing laser process parameters enable rapid and low-energy fabrication of SERS substrate. The functionalities including sensitivity, reproducibility, stability, and renewability are evaluated using rhodamine 6G as a probe molecule. Then, the feasibility of POCT is examined by the statistical analysis of SERS spectra of exosomes from malignant breast cancer cells and non-tumorigenic breast epithelial cells. The presented framework is anticipated to be utilized in other biomedical applications, facilitating cost-effective and large-scale production performance.
Collapse
Affiliation(s)
- Jong-Eun Park
- Department of Mechanical Engineering, The State University of New York, Korea (SUNY Korea), Incheon, 21985, Republic of Korea
| | - Hyeono Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - June Sik Hwang
- Department of Mechanical Engineering, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seong Jae Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sanha Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Minyang Yang
- Department of Mechanical Engineering, The State University of New York, Korea (SUNY Korea), Incheon, 21985, Republic of Korea
| |
Collapse
|
8
|
Qi G, Diao X, Tian Y, Sun D, Jin Y. Electroactivated SERS Nanoplatform for Rapid and Sensitive Detection and Identification of Tumor-Derived Exosome miRNA. Anal Chem 2024; 96:18519-18527. [PMID: 39523538 DOI: 10.1021/acs.analchem.4c04402] [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: 11/16/2024]
Abstract
Exosomal miRNA expression derived from tumor cells provides a valuable and promising noninvasive modality for the early diagnosis and assessment of the efficacy of cancer treatment. However, accurate detection and identification of miRNA within exosomes have been challenging due to its low abundance and the complexity and tedious extraction with large sample volumes in the separation process. Here, we developed an electrically activated nanoplatform for rapid and sensitive detection and identification of exosome miRNA, through triggering miRNA release by opening exosomes that were captured on the electrode surface using a slightly applied electric field (50 mV), and simultaneously detected them with surface-enhanced Raman spectroscopy (SERS) in situ. The method possessed superior specificity and sensitivity for exosomal miRNA detection, with a low detection concentration of 0.5 nM. The SERS sensor chips also showed a superior sensing performance of exosomal miRNA in complex body fluids such as urine and blood. We found that exosomal miRNA contents derived from tumor cells were significantly higher than those in normal cells, and importantly, the concentrations of exosomes secreted from three different cell lines were distinctly augmented after mild electrical stimulation (ES) treatment. Furthermore, the miRNA expression within exosomes was upregulated after the ES treatment of cells. The developed approach and SERS detection platform for exosomal miRNA are promising for noninvasive and precise screening, classification, and monitoring of cancer.
Collapse
Affiliation(s)
- Guohua Qi
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xingkang Diao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Yu Tian
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Dan Sun
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, P. R. China
| | - Yongdong Jin
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| |
Collapse
|
9
|
Sha A, Luo Y, Xiao W, He J, Chen X, Xiong Z, Peng L, Zou L, Liu B, Li Q. Plant-Derived Exosome-like Nanoparticles: A Comprehensive Overview of Their Composition, Biogenesis, Isolation, and Biological Applications. Int J Mol Sci 2024; 25:12092. [PMID: 39596159 PMCID: PMC11593521 DOI: 10.3390/ijms252212092] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/03/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Plant-derived exosome-like nanoparticles (PELNs) are a type of membranous vesicle isolated from plant tissues. They contain proteins, lipids, nucleic acids, and other components. PELNs are involved in the defensive response to pathogen attacks by exerting anti-inflammatory, antiviral, antifibrotic, and antitumor effects through the substances they contain. Most PELNs are edible and can be used as carriers for delivering specific drugs without toxicity and side effects, making them a hot topic of research. Sources of PELNs are abundantly, and they can be produced in high yields, with a low risk of developing immunogenicity in vivo. This paper summarizes the formation, isolation, and purification methods; physical properties; and composition of PELNs through a comprehensive literature search. It also analyzes the biomedical applications of PELNs, as well as future research directions. This paper provides new ideas and methods for future research on PELNs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Bingliang Liu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, School of Food and Biological Engineering, Chengdu University, No. 2025, Chengluo Avenue, Longquanyi District, Chengdu 610106, China; (A.S.); (Y.L.); (W.X.); (J.H.); (X.C.); (Z.X.); (L.P.); (L.Z.)
| | - Qiang Li
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, School of Food and Biological Engineering, Chengdu University, No. 2025, Chengluo Avenue, Longquanyi District, Chengdu 610106, China; (A.S.); (Y.L.); (W.X.); (J.H.); (X.C.); (Z.X.); (L.P.); (L.Z.)
| |
Collapse
|
10
|
Liu Z, Ng M, Srivastava S, Li T, Liu J, Phu TA, Mateescu B, Wang YT, Tsai CF, Liu T, Raffai RL, Xie YH. Label-free single-vesicle based surface enhanced Raman spectroscopy: A robust approach for investigating the biomolecular composition of small extracellular vesicles. PLoS One 2024; 19:e0305418. [PMID: 38889139 PMCID: PMC11185487 DOI: 10.1371/journal.pone.0305418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
Small extracellular vesicles (sEVs) are cell-released vesicles ranging from 30-150nm in size. They have garnered increasing attention because of their potential for both the diagnosis and treatment of disease. The diversity of sEVs derives from their biological composition and cargo content. Currently, the isolation of sEV subpopulations is primarily based on bio-physical and affinity-based approaches. Since a standardized definition for sEV subpopulations is yet to be fully established, it is important to further investigate the correlation between the biomolecular composition of sEVs and their physical properties. In this study, we employed a platform combining single-vesicle surface-enhanced Raman spectroscopy (SERS) and machine learning to examine individual sEVs isolated by size-exclusion chromatography (SEC). The biomolecular composition of each vesicle examined was reflected by its corresponding SERS spectral features (biomolecular "fingerprints"), with their roots in the composition of their collective Raman-active bonds. Origins of the SERS spectral features were validated through a comparative analysis between SERS and mass spectrometry (MS). SERS fingerprinting of individual vesicles was effective in overcoming the challenges posed by EV population averaging, allowing for the possibility of analyzing the variations in biomolecular composition between the vesicles of similar and/or different sizes. Using this approach, we uncovered that each of the size-based fractions of sEVs contained particles with predominantly similar SERS spectral features. Indeed, more than 84% of the vesicles residing within a particular group were clearly distinguishable from that of the other EV sub-populations, despite some spectral variations within each sub-population. Our results suggest the possibility that size-based EV fractionation methods produce samples where similarly eluted sEVs are correlated with their respective biochemical contents, as reflected by their SERS spectra. Our findings therefore highlight the possibility that the biogenesis and respective biological functionalities of the various sEV fractions may be inherently different.
Collapse
Affiliation(s)
- Zirui Liu
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, California, United States of America
| | - Martin Ng
- Northern California Institute for Research and Education, San Francisco, California, United States of America
| | - Siddharth Srivastava
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tieyi Li
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jun Liu
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tuan Anh Phu
- Northern California Institute for Research and Education, San Francisco, California, United States of America
| | - Bogdan Mateescu
- Brain Research Institute, University of Zürich, Zürich, Switzerland
- Institute for Chemical and Bioengineering, ETH Zürich, Zürich, Switzerland
| | - Yi-Ting Wang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, United States of America
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, United States of America
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, United States of America
| | - Robert L. Raffai
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, California, United States of America
| | - Ya-Hong Xie
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, California, United States of America
- UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
11
|
Lyu N, Hassanzadeh-Barforoushi A, Rey Gomez LM, Zhang W, Wang Y. SERS biosensors for liquid biopsy towards cancer diagnosis by detection of various circulating biomarkers: current progress and perspectives. NANO CONVERGENCE 2024; 11:22. [PMID: 38811455 PMCID: PMC11136937 DOI: 10.1186/s40580-024-00428-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024]
Abstract
Liquid biopsy has emerged as a promising non-invasive strategy for cancer diagnosis, enabling the detection of various circulating biomarkers, including circulating tumor cells (CTCs), circulating tumor nucleic acids (ctNAs), circulating tumor-derived small extracellular vesicles (sEVs), and circulating proteins. Surface-enhanced Raman scattering (SERS) biosensors have revolutionized liquid biopsy by offering sensitive and specific detection methodologies for these biomarkers. This review comprehensively examines the application of SERS-based biosensors for identification and analysis of various circulating biomarkers including CTCs, ctNAs, sEVs and proteins in liquid biopsy for cancer diagnosis. The discussion encompasses a diverse range of SERS biosensor platforms, including label-free SERS assay, magnetic bead-based SERS assay, microfluidic device-based SERS system, and paper-based SERS assay, each demonstrating unique capabilities in enhancing the sensitivity and specificity for detection of liquid biopsy cancer biomarkers. This review critically assesses the strengths, limitations, and future directions of SERS biosensors in liquid biopsy for cancer diagnosis.
Collapse
Affiliation(s)
- Nana Lyu
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | | | - Laura M Rey Gomez
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
12
|
Srivastava S, Sandhu N, Liu J, Xie YH. AI-Driven Spectral Decomposition: Predicting the Most Probable Protein Compositions from Surface Enhanced Raman Spectroscopy Spectra of Amino Acids. Bioengineering (Basel) 2024; 11:482. [PMID: 38790349 PMCID: PMC11117800 DOI: 10.3390/bioengineering11050482] [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: 03/03/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Surface-enhanced Raman spectroscopy (SERS) is a powerful tool for elucidating the molecular makeup of materials. It possesses the unique characteristics of single-molecule sensitivity and extremely high specificity. However, the true potential of SERS, particularly in capturing the biochemical content of particles, remains underexplored. In this study, we harnessed transformer neural networks to interpret SERS spectra, aiming to discern the amino acid profiles within proteins. By training the network on the SERS profiles of 20 amino acids of human proteins, we explore the feasibility of predicting the predominant proteins within the µL-scale detection volume of SERS. Our results highlight a consistent alignment between the model's predictions and the protein's known amino acid compositions, deepening our understanding of the inherent information contained within SERS spectra. For instance, the model achieved low root mean square error (RMSE) scores and minimal deviation in the prediction of amino acid compositions for proteins such as Bovine Serum Albumin (BSA), ACE2 protein, and CD63 antigen. This novel methodology offers a robust avenue not only for protein analytics but also sets a precedent for the broader realm of spectral analyses across diverse material categories. It represents a solid step forward to establishing SERS-based proteomics.
Collapse
Affiliation(s)
| | | | | | - Ya-Hong Xie
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA; (S.S.); (N.S.); (J.L.)
| |
Collapse
|
13
|
Zhang Q, Ren T, Cao K, Xu Z. Advances of machine learning-assisted small extracellular vesicles detection strategy. Biosens Bioelectron 2024; 251:116076. [PMID: 38340580 DOI: 10.1016/j.bios.2024.116076] [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: 09/05/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
Detection of extracellular vesicles (EVs), particularly small EVs (sEVs), is of great significance in exploring their physiological characteristics and clinical applications. The heterogeneity of sEVs plays a crucial role in distinguishing different types of cells and diseases. Machine learning, with its exceptional data processing capabilities, offers a solution to overcome the limitations of conventional detection methods for accurately classifying sEV subtypes and sources. Principal component analysis, linear discriminant analysis, partial least squares discriminant analysis, XGBoost, support vector machine, k-nearest neighbor, and deep learning, along with some combined methods such as principal component-linear discriminant analysis, have been successfully applied in the detection and identification of sEVs. This review focuses on machine learning-assisted detection strategies for cell identification and disease prediction via sEVs, and summarizes the integration of these strategies with surface-enhanced Raman scattering, electrochemistry, inductively coupled plasma mass spectrometry and fluorescence. The performance of different machine learning-based detection strategies is compared, and the advantages and limitations of various machine learning models are also evaluated. Finally, we discuss the merits and limitations of the current approaches and briefly outline the perspective of potential research directions in the field of sEV analysis based on machine learning.
Collapse
Affiliation(s)
- Qi Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Tingju Ren
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Ke Cao
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
14
|
Chen M, Wang H, Zhang Y, Jiang H, Li T, Liu L, Zhao Y. Label-Free Multiplex Profiling of Exosomal Proteins with a Deep Learning-Driven 3D Surround-Enhancing SERS Platform for Early Cancer Diagnosis. Anal Chem 2024; 96:6794-6801. [PMID: 38624007 DOI: 10.1021/acs.analchem.4c00669] [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: 04/17/2024]
Abstract
Identification of protein profiling on plasma exosomes by SERS can be a promising strategy for early cancer diagnosis. However, it is still challenging to detect multiple exosomal proteins simultaneously by SERS since the Raman signals of exosomes detected by conventional colloidal nanocrystals or two-dimensional SERS substrates are incomplete and complex. Herein, we develop a novel three-dimensional (3D) surround-enhancing SERS platform, named 3D se-SERS, for the multiplex detection of exosomal proteins. In this 3D se-SERS, proteins and exosomes are covered with "hotspots" generated by the gold nanoparticles, which surround the analytes densely and three-dimensionally, providing sensitive and comprehensive SERS signals. Combining this 3D se-SERS with a deep learning model, we successfully quantitatively profiled seven proteins including CD63, CD81, CD9, CD151, CD171, TSPAN8, and PD-L1 on the surface of plasma exosomes from patients, which can predict the occurrence and advancement of lung cancer. This 3D se-SERS integrating deep learning technique benefits from high sensitivity and significant multiplexing ability for comprehensive analysis of proteins and exosomes, demonstrating the potential of deep learning-driven 3D se-SERS technology for plasma exosome-based early cancer diagnosis.
Collapse
Affiliation(s)
- Miao Chen
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Haoyang Wang
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Yibin Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hanyu Jiang
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Tan Li
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Lixin Liu
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Yuetao Zhao
- School of Life Sciences, Central South University, Changsha 410013, China
| |
Collapse
|
15
|
Shen J, Ma Z, Xu J, Xue T, Lv X, Zhu G, Huang B. Exosome Isolation and Detection: From Microfluidic Chips to Nanoplasmonic Biosensor. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38676635 DOI: 10.1021/acsami.3c19396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Exosomes are becoming more widely acknowledged as significant circulating indicators for the prognosis and diagnosis of cancer. Circulating exosomes are essential to the development and spread of cancer, according to a growing body of research. Using existing technology, characterizing exosomes is quite difficult. Therefore, a direct, sensitive, and targeted approach to exosome detection will aid in illness diagnosis and prognosis. The review discusses the new strategies for exosome isolation and detection technologies from microfluidic chips to nanoplasmonic biosensors, analyzing the advantages and limitations of these new technologies. This review serves researchers to better understand exosome isolation and detection methods and to help develop better exosome isolating and detecting devices for clinical applications.
Collapse
Affiliation(s)
- Jianing Shen
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Zhengtai Ma
- Key Laboratory of Optoelectronic Materials and Devices, Chinese Academy of Sciences, Beijing 100083, China
- College of Materials Science and Optoelectronic Technology, University of Chinese, Academy of Sciences, Beijing 100049, China
| | - Jiaqi Xu
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Tianhao Xue
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Xiaoqing Lv
- Key Laboratory of Optoelectronic Materials and Devices, Chinese Academy of Sciences, Beijing 100083, China
| | - Guixian Zhu
- School of Instrument Science and Optoelectronic Engineering, Beijing Information Science and Technology University, Beijing 100192, China
| | - Beiju Huang
- Key Laboratory of Optoelectronic Materials and Devices, Chinese Academy of Sciences, Beijing 100083, China
- College of Materials Science and Optoelectronic Technology, University of Chinese, Academy of Sciences, Beijing 100049, China
| |
Collapse
|
16
|
Buccini L, Proietti A, La Penna G, Mancini C, Mura F, Tacconi S, Dini L, Rossi M, Passeri D. Toward the nanoscale chemical and physical probing of milk-derived extracellular vesicles using Raman and tip-enhanced Raman spectroscopy. NANOSCALE 2024; 16:8132-8142. [PMID: 38568015 DOI: 10.1039/d4nr00845f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Tip-enhanced Raman spectroscopy (TERS) is an advanced technique to perform local chemical analysis of the surface of a sample through the improvement of the sensitivity and the spatial resolution of Raman spectroscopy by plasmonic enhancement of the electromagnetic signal in correspondence with the nanometer-sized tip of an atomic force microscope (AFM). In this work, TERS is demonstrated to represent an innovative and powerful approach for studying extracellular vesicles, in particular bovine milk-derived extracellular vesicles (mEVs), which are nanostructures with considerable potential in drug delivery and therapeutic applications. Raman spectroscopy has been used to analyze mEVs at the micrometric and sub-micrometric scales to obtain a detailed Raman spectrum in order to identify the 'signature' of mEVs in terms of their characteristic molecular vibrations and, therefore, their chemical compositions. With the ability to improve lateral resolution, TERS has been used to study individual mEVs, demonstrating the possibility of investigating a single mEV selected on the surface of the sample and, moreover, analyzing specific locations on the selected mEV with nanometer lateral resolution. TERS potentially allows one to reveal local differences in the composition of mEVs providing new insights into their structure. Also, thanks to the intrinsic properties of TERS to acquire the signal from only the first few nanometers of the surface, chemical investigation of the lipid membrane in correspondence with the various locations of the selected mEV could be performed by analyzing the peaks of the Raman shift in the relevant range of the spectrum (2800-3000 cm-1). Despite being limited to mEVs, this work demonstrates the potential of TERS in the analysis of extracellular vesicles.
Collapse
Affiliation(s)
- Luca Buccini
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Anacleto Proietti
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Giancarlo La Penna
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Chiara Mancini
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Francesco Mura
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
- Research Center for Nanotechnology Applied to Engineering of Sapienza University of Rome (CNIS), Piazzale A. Moro 5, 00185 Rome, Italy
| | - Stefano Tacconi
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Luciana Dini
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Marco Rossi
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
- Research Center for Nanotechnology Applied to Engineering of Sapienza University of Rome (CNIS), Piazzale A. Moro 5, 00185 Rome, Italy
| | - Daniele Passeri
- Department of Basic and Applied Sciences for Engineering, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
- Research Center for Nanotechnology Applied to Engineering of Sapienza University of Rome (CNIS), Piazzale A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
17
|
Tang W, Zhao K, Li X, Zhou X, Liao P. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Promote the Recovery of Spinal Cord Injury and Inhibit Ferroptosis by Inactivating IL-17 Pathway. J Mol Neurosci 2024; 74:33. [PMID: 38536541 DOI: 10.1007/s12031-024-02209-3] [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: 01/23/2024] [Accepted: 03/16/2024] [Indexed: 04/11/2024]
Abstract
Mesenchymal stem cell (MSC)-derived exosomes are considered as alternative to cell therapy in various diseases. This study aimed to understand the effect of bone marrow MSC-derived exosomes (BMMSC-exos) on spinal cord injury (SCI) and to unveil its regulatory mechanism on ferroptosis. Exosomes were isolated from BMMSCs and the uptake of BMMSCs-exos by PC12 cells was determined using PKH67 staining. The effect of BMMSC-exos on SCI in rats was studied by evaluating pathological changes of spinal cord tissues, inflammatory cytokines, and ferroptosis-related proteins. Transcriptome sequencing was used to discover the differential expressed genes (DEGs) between SCI rats and BMMSC-exos-treated rats followed by functional enrichment analyses. The effect of BMMSC-exos on ferroptosis and interleukin 17 (IL-17) pathway was evaluated in SCI rats and oxygen-glucose deprivation (OGD)-treated PC12 cells. The results showed that particles extracted from BMMSCs were exosomes that could be taken up by PC12 cells. BMMSC-exos treatment ameliorated injuries of spinal cord, suppressed the accumulation of Fe2+, malondialdehyde (MDA), and reactive oxygen species (ROS), with the elevated glutathione (GSH). Also, BMMSC-exos downregulated the expression of acyl-CoA synthetase long chain family member 4 (ACSL4) and upregulated glutathione peroxidase 4 (GPX4) and cysteine/glutamate antiporter xCT. A total of 110 DEGs were discovered and they were mainly enriched in IL-17 signaling pathway. Further in vitro and in vivo experiments showed that BMMSC-exos inactivated IL-17 pathway. BMMSC-exos promote the recovery of SCI and inhibit ferroptosis by inhibiting the IL-17 pathway, which provides BMMSC-exos as an alternative to the management of SCI.
Collapse
Affiliation(s)
- Wen Tang
- Department of Trauma Center, The First Affiliated Hospital of Gannan Medical University, No. 128, West Jinling Road, Ganzhou, 341000, China.
| | - Kai Zhao
- Department of Spine Surgery, The First Affiliated Hospital of Gannan Medical University, No. 128, West Jinling Road, Ganzhou, 341000, China
| | - Xiaobo Li
- Center for Technology of Information and Network Management, Gannan Medical University, Ganzhou, 341000, China
| | - Xiaozhong Zhou
- Department of Trauma Center, The First Affiliated Hospital of Gannan Medical University, No. 128, West Jinling Road, Ganzhou, 341000, China
| | - Peigen Liao
- The First Clinical Medical College, Gannan Medical University, No. 128, West Jinling Road, Ganzhou, 341000, China
| |
Collapse
|
18
|
Han J, Jung JH, Lee SY, Park JH. Nanoplasmonic Detection of EGFR Mutations Based on Extracellular Vesicle-Derived EGFR-Drug Interaction. ACS APPLIED MATERIALS & INTERFACES 2024; 16:8266-8274. [PMID: 38335730 DOI: 10.1021/acsami.3c14907] [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: 02/12/2024]
Abstract
Analysis of membrane proteins from extracellular vesicles (EVs) has emerged as an important strategy for molecular cancer diagnosis. The epidermal growth factor receptor (EGFR) is one of the most well-known oncogenic membrane proteins, particularly in non-small cell lung cancer (NSCLC), where targeted therapies using tyrosine kinase inhibitors (TKIs) are often addressed based on EGFR mutation status. Consequently, several studies aimed at analyzing oncogenic membrane proteins have been proposed for cancer diagnosis. However, conventional protein analysis still faces limitations due to the requirement for large sample quantities and extensive post-labeling processes. Here, we develop a nanoplasmonic detection method for EGFR mutations in the diagnosis of NSCLC based on interactions between EGFR loaded in EVs and TKI. Gefitinib is selected as a model TKI due to its strong signals in the surface-enhanced Raman spectroscopy (SERS) and mutation-dependent binding affinity to EGFR. We demonstrate an SERS signal attributed to gefitinib at a higher value in the EGFR exon 19 deletion, both in cells and EVs, compared to wild-type and exon 19 deletion/T790M variants. Furthermore, we observe a significantly higher gefitinib SERS signal in EGFR obtained from exon 19 deletion NSCLC patient plasma-derived EVs compared with those from wild-type and exon 19 deletion/T790M EVs. Since our approach utilizes an analysis of the SERS signal generated by the interaction between oncogenic membrane proteins within EVs and targeted drugs, its diagnostic applicability could potentially extend to other liquid biopsy methods based on EVs.
Collapse
Affiliation(s)
- Junhee Han
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jik-Han Jung
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Sung Yong Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
19
|
Cong XZ, Feng J, Zhang HJ, Zhang LZ, Lin TY, Chen G, Zhang ZL. Microfluidic Device-Based In Vivo Detection of PD-L1-Positive Small Extracellular Vesicles and Its Application for Tumor Monitoring. Anal Chem 2024; 96:2658-2665. [PMID: 38311857 DOI: 10.1021/acs.analchem.3c05418] [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: 02/06/2024]
Abstract
Liquid biopsy is of great significance in tumor early diagnosis and treatment stratification. PD-L1-positive small extracellular vesicles (PD-L1+ sEVs) are closely related to tumor growth and immunotherapy response, which are considered valuable liquid biopsy biomarkers. In contrast to conventional in vitro detection, in vivo detection has the ability to improve the detection efficiency and enable continuous or real-time dynamic monitoring. However, in vivo detection of PD-L1+ sEVs has multiple difficulties, such as high cell background, complex blood environments, and lack of a specific and stable detection method. Herein, the in vivo detection of PD-L1+ sEVs method was constructed, which efficiently separated sEVs based on the microfluidic device and quantitatively analyzed PD-L1+ sEVs by aptamer recognition and hybridization chain reaction. The concentration of PD-L1+ sEVs was continuously monitored, and significant differences at different stages of tumor as well as a correlation with tumor volume were found. Diseased and healthy individuals could also be effectively distinguished based on the concentration of PD-L1+ sEVs. The method with good stability, biocompatibility, and detection performance provided a powerful means for in vivo detection of PD-L1+ sEVs, contributing to the clinical diagnosis and treatment of tumor.
Collapse
Affiliation(s)
- Xi-Zhu Cong
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Jiao Feng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - He-Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Lin-Zhou Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Tian-Yang Lin
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
20
|
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
|
21
|
Srivastava S, Terai Y, Liu J, Capellini G, Xie YH. Controlling the Nucleation and Growth of Salt from Bodily Fluid for Enhanced Biosensing Applications. BIOSENSORS 2023; 13:1016. [PMID: 38131777 PMCID: PMC10741434 DOI: 10.3390/bios13121016] [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: 11/16/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Surface-enhanced Raman spectroscopy (SERS) represents a transformative tool in medical diagnostics, particularly for the early detection of key biomarkers such as small extracellular vesicles (sEVs). Its unparalleled sensitivity and compatibility with intricate biological samples make it an ideal candidate for revolutionizing noninvasive diagnostic methods. However, a significant challenge that mars its efficacy is the throughput limitation, primarily anchored in the prerequisite of hotspot and sEV colocalization within a minuscule range. This paper delves deep into this issue, introducing a never-attempted-before approach which harnesses the principles of crystallization-nucleation and growth. By synergistically coupling lasers with plasmonic resonances, we navigate the challenges associated with the analyte droplet drying method and the notorious coffee ring effect. Our method, rooted in a profound understanding of crystallization's materials science, exhibits the potential to significantly increase the areal density of accessible plasmonic hotspots and efficiently guide exosomes to defined regions. In doing so, we not only overcome the throughput challenge but also promise a paradigm shift in the arena of minimally invasive biosensing, ushering in advanced diagnostic capabilities for life-threatening diseases.
Collapse
Affiliation(s)
- Siddharth Srivastava
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA
| | - Yusuke Terai
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya 464-8601, Japan
| | - Jun Liu
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA
| | - Giovanni Capellini
- IHP—Leibniz Institute for High Performance Microelectronics, 15236 Frankfurt (Oder), Germany;
- Department of Science, Università Degli Studi Roma Tre, Viale Marconi 446, 00146 Rome, Italy
| | - Ya-Hong Xie
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
22
|
Taha HB. Plasma versus serum for extracellular vesicle (EV) isolation: A duel for reproducibility and accuracy for CNS-originating EVs biomarker analysis. J Neurosci Res 2023; 101:1677-1686. [PMID: 37501394 DOI: 10.1002/jnr.25231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023]
Abstract
Blood-derived extracellular vesicles (EVs) are a popular source of biomarkers for central nervous system (CNS) diseases, but inconsistencies in isolation and analysis hinder their clinical translation. This review summarizes recent studies that investigate the impact of different anticoagulated plasma and serum on the yield, purity, and molecular content of EVs. Specifically, the studies compare ethylenediaminetetraacetic acid (EDTA), citrate, heparin plasma, and serum and highlight the risk of contamination from platelet-derived EVs. Here, I offer practical guidelines for standardizing EV isolation and analysis, recommending the use of plasma anticoagulated with acid-citrate-dextrose (ACD) or citrate followed by EDTA and heparin, subgroup analyses for samples from different biobank repositories, and avoiding serum and plasma-to-serum transformation. Other factors like illness, age, gender, meal timing, exercise, circadian timing, and arm pressure during blood draw can alter EV signatures. Yet, how these variables interact with different anticoagulated plasma or serum samples is unclear, necessitating further research. Furthermore, whether the changes are dependent on the isolation or quantification methodology remains an area of investigation. Importantly, the perspective emphasizes the need for consistency in experimental methodologies to improve the reproducibility and clinical applicability of CNS-originating EV biomarker studies. The proposed guidelines, along with ongoing efforts to standardize blood sample handling and collection, may facilitate the development of more reliable and informative CNS-originating EV biomarkers for diagnosis, prognosis, and treatment monitoring of CNS diseases.
Collapse
Affiliation(s)
- Hash Brown Taha
- Department of Integrative Biology & Physiology, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
23
|
Premachandran S, Haldavnekar R, Ganesh S, Das S, Venkatakrishnan K, Tan B. Self-Functionalized Superlattice Nanosensor Enables Glioblastoma Diagnosis Using Liquid Biopsy. ACS NANO 2023; 17:19832-19852. [PMID: 37824714 DOI: 10.1021/acsnano.3c04118] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Glioblastoma (GBM), the most aggressive and lethal brain cancer, is detected only in the advanced stage, resulting in a median survival rate of 15 months. Therefore, there is an urgent need to establish GBM diagnosis tools to identify the tumor accurately. The clinical relevance of the current liquid biopsy techniques for GBM diagnosis remains mostly undetermined, owing to the challenges posed by the blood-brain barrier (BBB) that restricts biomarkers entering the circulation, resulting in the unavailability of clinically validated circulating GBM markers. GBM-specific liquid biopsy for diagnosis and prognosis of GBM has not yet been developed. Here, we introduce extracellular vesicles of GBM cancer stem cells (GBM CSC-EVs) as a previously unattempted, stand-alone GBM diagnosis modality. As GBM CSCs are fundamental building blocks of tumor initiation and recurrence, it is desirable to investigate these reliable signals of malignancy in circulation for accurate GBM diagnosis. So far, there are no clinically validated circulating biomarkers available for GBM. Therefore, a marker-free approach was essential since conventional liquid biopsy relying on isolation methodology was not viable. Additionally, a mechanism capable of trace-level detection was crucial to detecting the rare GBM CSC-EVs from the complex environment in circulation. To break these barriers, we applied an ultrasensitive superlattice sensor, self-functionalized for surface-enhanced Raman scattering (SERS), to obtain holistic molecular profiling of GBM CSC-EVs with a marker-free approach. The superlattice sensor exhibited substantial SERS enhancement and ultralow limit of detection (LOD of attomolar 10-18 M concentration) essential for trace-level detection of invisible GBM CSC-EVs directly from patient serum (without isolation). We detected as low as 5 EVs in 5 μL of solution, achieving the lowest LOD compared to existing SERS-based studies. We have experimentally demonstrated the crucial role of the signals of GBM CSC-EVs in the precise detection of glioblastoma. This was evident from the unique molecular profiles of GBM CSC-EVs demonstrating significant variation compared to noncancer EVs and EVs of GBM cancer cells, thus adding more clarity to the current understanding of GBM CSC-EVs. Preliminary validation of our approach was undertaken with a small amount of peripheral blood (5 μL) derived from GBM patients with 100% sensitivity and 97% specificity. Identification of the signals of GBM CSC-EV in clinical sera specimens demonstrated that our technology could be used for accurate GBM detection. Our technology has the potential to improve GBM liquid biopsy, including real-time surveillance of GBM evolution in patients upon clinical validation. This demonstration of liquid biopsy with GBM CSC-EV provides an opportunity to introduce a paradigm potentially impacting the current landscape of GBM diagnosis.
Collapse
Affiliation(s)
- Srilakshmi Premachandran
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Rupa Haldavnekar
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Swarna Ganesh
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Sunit Das
- Scientist, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Institute of Medical Sciences, Neurosurgery, University of Toronto, Toronto, Ontario M5T 1P5, Canada
| | - Krishnan Venkatakrishnan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Bo Tan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University (formerly Ryerson University) and St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
- Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University (formerly Ryerson University), 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| |
Collapse
|
24
|
Nam H, Park JE, Waheed W, Alazzam A, Sung HJ, Jeon JS. Acoustofluidic lysis of cancer cells and Raman spectrum profiling. LAB ON A CHIP 2023; 23:4117-4125. [PMID: 37655531 DOI: 10.1039/d3lc00550j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
The lysis of cancer cells inside a sessile droplet was performed using traveling surface acoustic waves (SAWs) without any chemical reagents. Raman spectrum profiling was then carried out to explore detailed cell-derived data. The Rayleigh waves formed by an interdigital transducer were made to propagate along the surface of an LiNbO3 substrate. Polystyrene microparticles (PSMPs) were used to establish mechanical cell lysis effectively, and gold nanoparticles (AuNPs) were added to enhance the Raman signals from the lysed cells by SAWs. The lysis efficiency was evaluated according to the size and concentration of the PSMPs in experiments where the frequency was varied. Lysis occurred mainly by mechanical collision using PSMPs in a high-frequency domain, and the lysis efficiency was improved by increasing the application time and the energy density of the SAWs. Raman signals from the lysed cells were greatly enhanced by nanogaps formed by the AuNPs, which were evenly distributed irrespective of the SAWs through the frequency-independent behavior of the AuNPs. Finally, detailed Raman spectra of MDA-MB-231, malignant breast cancer cells, were acquired, and various organic matter-derived peaks were observed. The 95% confidence region for cells subjected to lysis was more widely distributed than that of cells not subjected to lysis. The proposed SAW platform is expected to facilitate the detection of small quantities and to be applied in biomedical applications.
Collapse
Affiliation(s)
- Hyeono Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Jong-Eun Park
- Department of Mechanical Engineering, The State University of New York Korea, Incheon 21985, Republic of Korea
| | - Waqas Waheed
- Department of Mechanical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Anas Alazzam
- Department of Mechanical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Hyung Jin Sung
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
25
|
Liu X, Xiao C, Xiao K. Engineered extracellular vesicles-like biomimetic nanoparticles as an emerging platform for targeted cancer therapy. J Nanobiotechnology 2023; 21:287. [PMID: 37608298 PMCID: PMC10463632 DOI: 10.1186/s12951-023-02064-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
Nanotechnology offers the possibility of revolutionizing cancer theranostics in the new era of precision oncology. Extracellular vesicles (EVs)-like biomimetic nanoparticles (EBPs) have recently emerged as a promising platform for targeted cancer drug delivery. Compared with conventional synthetic vehicles, EBPs have several advantages, such as lower immunogenicity, longer circulation time, and better targeting capability. Studies on EBPs as cancer therapeutics are rapidly progressing from in vitro experiments to in vivo animal models and early-stage clinical trials. Here, we describe engineering strategies to further improve EBPs as effective anticancer drug carriers, including genetic manipulation of original cells, fusion with synthetic nanomaterials, and direct modification of EVs. These engineering approaches can improve the anticancer performance of EBPs, especially in terms of tumor targeting effectiveness, stealth property, drug loading capacity, and integration with other therapeutic modalities. Finally, the current obstacles and future perspectives of engineered EBPs as the next-generation delivery platform for anticancer drugs are discussed.
Collapse
Affiliation(s)
- Xinyi Liu
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunxiu Xiao
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Xiao
- Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, Frontiers Science Center for Disease-Related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Tianfu Jingcheng Laboratory (Frontier Medical Center), Chengdu, 610041, China.
| |
Collapse
|
26
|
Kaur S, Nathani A, Singh M. Exosomal delivery of cannabinoids against cancer. Cancer Lett 2023; 566:216243. [PMID: 37257632 PMCID: PMC10426019 DOI: 10.1016/j.canlet.2023.216243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/11/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
Exosomes are extracellular vesicles (EVs) originating from endosomes that play a role in cellular communication. These vesicles which mimic the parental cells that release them are promising candidates for targeted drug delivery and therapeutic applications against cancer because of their favorable biocompatibility, specific targeting, low toxicity, and immunogenicity. Currently, Delta-9-tetrahydrocannabinol (THC), cannabidiol (CBD) and other cannabinoids (e.g., CBG, THCV, CBC), are being explored for their anticancer and anti-proliferative properties. Several mechanisms, including cell cycle arrest, proliferation inhibition, activation of autophagy and apoptosis, inhibition of adhesion, metastasis, and angiogenesis have been proposed for their anticancer activity. EVs could be engineered as cannabinoid delivery systems for tumor-specificity leading to superior anticancer effects. This review discusses current techniques for EV isolation from various sources, characterization and strategies to load them with cannabinoids. More extensively, we culminate information available on different sources of EVs that have anticancer activity, mechanism of action of cannabinoids against various wild type and resistant tumors and role of CBD in histone modifications and cancer epigenetics. We have also enumerated the role of EVs containing cannabinoids against various tumors and in chemotherapy induced neuropathic pain.
Collapse
Affiliation(s)
- Sukhmandeep Kaur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA.
| |
Collapse
|
27
|
Tang Y, Liu X, Sun M, Xiong S, Xiao N, Li J, He X, Xie J. Recent Progress in Extracellular Vesicle-Based Carriers for Targeted Drug Delivery in Cancer Therapy. Pharmaceutics 2023; 15:1902. [PMID: 37514088 PMCID: PMC10384044 DOI: 10.3390/pharmaceutics15071902] [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: 05/11/2023] [Revised: 06/19/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Extracellular vesicles (EVs) are small, membrane-based vesicles released by cells that play a critical role in various physiological and pathological processes. They act as vehicles for transporting a variety of endogenous cargo molecules, enabling intercellular communication. Due to their natural properties, EVs have emerged as a promising "cell-free therapy" strategy for treating various diseases, including cancer. They serve as excellent carriers for different therapeutics, including nucleic acids, proteins, small molecules, and other nanomaterials. Modifying or engineering EVs can improve the efficacy, targeting, specificity, and biocompatibility of EV-based therapeutics for cancer therapy. In this review, we comprehensively outline the biogenesis, isolation, and methodologies of EVs, as well as their biological functions. We then focus on specific applications of EVs as drug carriers in cancer therapy by citing prominent recent studies. Additionally, we discuss the opportunities and challenges for using EVs as pharmaceutical drug delivery vehicles. Ultimately, we aim to provide theoretical and technical support for the development of EV-based carriers for cancer treatment.
Collapse
Affiliation(s)
- Yaqin Tang
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xingyou Liu
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Meng Sun
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Su Xiong
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Nianting Xiao
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Jianchao Li
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Xiao He
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Jing Xie
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
28
|
Gupta S, Dutta S, Hui SP. Regenerative Potential of Injured Spinal Cord in the Light of Epigenetic Regulation and Modulation. Cells 2023; 12:1694. [PMID: 37443728 PMCID: PMC10341208 DOI: 10.3390/cells12131694] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 07/15/2023] Open
Abstract
A spinal cord injury is a form of physical harm imposed on the spinal cord that causes disability and, in many cases, leads to permanent mammalian paralysis, which causes a disastrous global issue. Because of its non-regenerative aspect, restoring the spinal cord's role remains one of the most daunting tasks. By comparison, the remarkable regenerative ability of some regeneration-competent species, such as some Urodeles (Axolotl), Xenopus, and some teleost fishes, enables maximum functional recovery, even after complete spinal cord transection. During the last two decades of intensive research, significant progress has been made in understanding both regenerative cells' origins and the molecular signaling mechanisms underlying the regeneration and reconstruction of damaged spinal cords in regenerating organisms and mammals, respectively. Epigenetic control has gradually moved into the center stage of this research field, which has been helped by comprehensive work demonstrating that DNA methylation, histone modifications, and microRNAs are important for the regeneration of the spinal cord. In this review, we concentrate primarily on providing a comparison of the epigenetic mechanisms in spinal cord injuries between non-regenerating and regenerating species. In addition, we further discuss the epigenetic mediators that underlie the development of a regeneration-permissive environment following injury in regeneration-competent animals and how such mediators may be implicated in optimizing treatment outcomes for spinal cord injurie in higher-order mammals. Finally, we briefly discuss the role of extracellular vesicles (EVs) in the context of spinal cord injury and their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Samudra Gupta
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
| | - Subhra Prakash Hui
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| |
Collapse
|
29
|
Faur CI, Dinu C, Toma V, Jurj A, Mărginean R, Onaciu A, Roman RC, Culic C, Chirilă M, Rotar H, Fălămaș A, Știufiuc GF, Hedeșiu M, Almășan O, Știufiuc RI. A New Detection Method of Oral and Oropharyngeal Squamous Cell Carcinoma Based on Multivariate Analysis of Surface Enhanced Raman Spectra of Salivary Exosomes. J Pers Med 2023; 13:jpm13050762. [PMID: 37240933 DOI: 10.3390/jpm13050762] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Raman spectroscopy recently proved a tremendous capacity to identify disease-specific markers in various (bio)samples being a non-invasive, rapid, and reliable method for cancer detection. In this study, we first aimed to record vibrational spectra of salivary exosomes isolated from oral and oropharyngeal squamous cell carcinoma patients and healthy controls using surface enhancement Raman spectroscopy (SERS). Then, we assessed this method's capacity to discriminate between malignant and non-malignant samples by means of principal component-linear discriminant analysis (PC-LDA) and we used area under the receiver operating characteristics with illustration as the area under the curve to measure the power of salivary exosomes SERS spectra analysis to identify cancer presence. The vibrational spectra were collected on a solid plasmonic substrate developed in our group, synthesized using tangential flow filtered and concentrated silver nanoparticles, capable of generating very reproducible spectra for a whole range of bioanalytes. SERS examination identified interesting variations in the vibrational bands assigned to thiocyanate, proteins, and nucleic acids between the saliva of cancer and control groups. Chemometric analysis indicated discrimination sensitivity between the two groups up to 79.3%. The sensitivity is influenced by the spectral interval used for the multivariate analysis, being lower (75.9%) when the full-range spectra were used.
Collapse
Affiliation(s)
- Cosmin Ioan Faur
- Department of Oral Radiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Cristian Dinu
- Department of Maxillofacial Surgery and Implantology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Valentin Toma
- MedFuture-Research Center for Advanced Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Anca Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Radu Mărginean
- MedFuture-Research Center for Advanced Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Anca Onaciu
- MedFuture-Research Center for Advanced Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Rareș Călin Roman
- Department of Oral and Craniomaxillofacial Surgery, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Carina Culic
- Department of Odontology, Endodontics, Oral Pathology, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Magdalena Chirilă
- Department of Otorhinolaryngology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Horațiu Rotar
- Department of Oral and Craniomaxillofacial Surgery, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Alexandra Fălămaș
- Department of Molecular and Biomolecular Physics, National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | | | - Mihaela Hedeșiu
- Department of Oral Radiology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Oana Almășan
- Department of Prosthodontics and Dental Materials, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Rares Ionuț Știufiuc
- Department of Maxillofacial Surgery and Implantology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Department of Pharmaceutical Physics & Biophysics, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| |
Collapse
|
30
|
Dutta S, Hornung S, Taha HB, Bitan G. Biomarkers for parkinsonian disorders in CNS-originating EVs: promise and challenges. Acta Neuropathol 2023; 145:515-540. [PMID: 37012443 PMCID: PMC10071251 DOI: 10.1007/s00401-023-02557-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023]
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and oncosomes, are nano-sized particles enclosed by a lipid bilayer. EVs are released by virtually all eukaryotic cells and have been shown to contribute to intercellular communication by transporting proteins, lipids, and nucleic acids. In the context of neurodegenerative diseases, EVs may carry toxic, misfolded forms of amyloidogenic proteins and facilitate their spread to recipient cells in the central nervous system (CNS). CNS-originating EVs can cross the blood-brain barrier into the bloodstream and may be found in other body fluids, including saliva, tears, and urine. EVs originating in the CNS represent an attractive source of biomarkers for neurodegenerative diseases, because they contain cell- and cell state-specific biological materials. In recent years, multiple papers have reported the use of this strategy for identification and quantitation of biomarkers for neurodegenerative diseases, including Parkinson's disease and atypical parkinsonian disorders. However, certain technical issues have yet to be standardized, such as the best surface markers for isolation of cell type-specific EVs and validating the cellular origin of the EVs. Here, we review recent research using CNS-originating EVs for biomarker studies, primarily in parkinsonian disorders, highlight technical challenges, and propose strategies for overcoming them.
Collapse
Affiliation(s)
- Suman Dutta
- International Institute of Innovation and Technology, New Town, Kolkata, India
| | - Simon Hornung
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hash Brown Taha
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, 635 Charles E. Young Drive South/Gordon 451, Los Angeles, CA, 90095, USA
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, 635 Charles E. Young Drive South/Gordon 451, Los Angeles, CA, 90095, USA.
- Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Shin H, Choi BH, Shim O, Kim J, Park Y, Cho SK, Kim HK, Choi Y. Single test-based diagnosis of multiple cancer types using Exosome-SERS-AI for early stage cancers. Nat Commun 2023; 14:1644. [PMID: 36964142 PMCID: PMC10039041 DOI: 10.1038/s41467-023-37403-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/16/2023] [Indexed: 03/26/2023] Open
Abstract
Early cancer detection has significant clinical value, but there remains no single method that can comprehensively identify multiple types of early-stage cancer. Here, we report the diagnostic accuracy of simultaneous detection of 6 types of early-stage cancers (lung, breast, colon, liver, pancreas, and stomach) by analyzing surface-enhanced Raman spectroscopy profiles of exosomes using artificial intelligence in a retrospective study design. It includes classification models that recognize signal patterns of plasma exosomes to identify both their presence and tissues of origin. Using 520 test samples, our system identified cancer presence with an area under the curve value of 0.970. Moreover, the system classified the tumor organ type of 278 early-stage cancer patients with a mean area under the curve of 0.945. The final integrated decision model showed a sensitivity of 90.2% at a specificity of 94.4% while predicting the tumor organ of 72% of positive patients. Since our method utilizes a non-specific analysis of Raman signatures, its diagnostic scope could potentially be expanded to include other diseases.
Collapse
Affiliation(s)
- Hyunku Shin
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
- Korea Artificial Organ Center, Korea University, Seoul, 02841, Republic of Korea
| | - On Shim
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Jihee Kim
- EXoPERT Corporation, Seoul, 02580, Republic of Korea
| | - Yong Park
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Suk Ki Cho
- Division of Thoracic Surgery, Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
- Department of Biomedical Sciences, College of Medicine, Korea University, 02841, Seoul, Republic of Korea.
| | - Yeonho Choi
- EXoPERT Corporation, Seoul, 02580, Republic of Korea.
- School of Biomedical Engineering, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Engineering, Korea University, Seoul, 02841, Republic of Korea.
- Interdisciplinary Program in Precision Public Health, Korea University, 02841, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Abe C, Bhaswant M, Miyazawa T, Miyazawa T. The Potential Use of Exosomes in Anti-Cancer Effect Induced by Polarized Macrophages. Pharmaceutics 2023; 15:pharmaceutics15031024. [PMID: 36986884 PMCID: PMC10054161 DOI: 10.3390/pharmaceutics15031024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
The rapid development of aberrant cells outgrowing their normal bounds, which can subsequently infect other body parts and spread to other organs-a process known as metastasis-is one of the significant characteristics of cancer. The main reason why cancer patients die is because of widespread metastases. This abnormal cell proliferation varies in cancers of over a hundred types, and their response to treatment can vary substantially. Several anti-cancer drugs have been discovered to treat various tumors, yet they still have harmful side-effects. Finding novel, highly efficient targeted therapies based on modifications in the molecular biology of tumor cells is essential to reduce the indiscriminate destruction of healthy cells. Exosomes, an extracellular vesicle, are promising as a drug carrier for cancer therapy due to their good tolerance in the body. In addition, the tumor microenvironment is a potential target to regulate in cancer treatment. Therefore, macrophages are polarized toward M1 and M2 phenotypes, which are involved in cancer proliferation and are malignant. It is evident from recent studies that controlled macrophage polarization might contribute to cancer treatment, by the direct way of using miRNA. This review provides an insight into the potential use of exosomes to develop an 'indirect', more natural, and harmless cancer treatment through regulating macrophage polarization.
Collapse
Affiliation(s)
- Chizumi Abe
- New Industry Creation Hatchery Center (NICHe), Tohoku University, 6-6-10 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - Maharshi Bhaswant
- New Industry Creation Hatchery Center (NICHe), Tohoku University, 6-6-10 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - Teruo Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, 6-6-10 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - Taiki Miyazawa
- New Industry Creation Hatchery Center (NICHe), Tohoku University, 6-6-10 Aramaki-aza-Aoba, Aoba-ku, Sendai 980-8579, Japan
| |
Collapse
|
33
|
Parlatan U, Ozen MO, Kecoglu I, Koyuncu B, Torun H, Khalafkhany D, Loc I, Ogut MG, Inci F, Akin D, Solaroglu I, Ozoren N, Unlu MB, Demirci U. Label-Free Identification of Exosomes using Raman Spectroscopy and Machine Learning. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205519. [PMID: 36642804 DOI: 10.1002/smll.202205519] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Exosomes, nano-sized extracellular vesicles (EVs) secreted from cells, carry various cargo molecules reflecting their cells of origin. As EV content, structure, and size are highly heterogeneous, their classification via cargo molecules by determining their origin is challenging. Here, a method is presented combining surface-enhanced Raman spectroscopy (SERS) with machine learning algorithms to employ the classification of EVs derived from five different cell lines to reveal their cellular origins. Using an artificial neural network algorithm, it is shown that the label-free Raman spectroscopy method's prediction ratio correlates with the ratio of HT-1080 exosomes in the mixture. This machine learning-assisted SERS method enables a new direction through label-free investigation of EV preparations by differentiating cancer cell-derived exosomes from those of healthy. This approach will potentially open up new avenues of research for early detection and monitoring of various diseases, including cancer.
Collapse
Affiliation(s)
- Ugur Parlatan
- Department of Physics, Bogazici University, Istanbul, 34342, Turkey
- Department of Radiology Stanford School of Medicine, BioAcoustic MEMS in Medicine Lab (BAMM), Canary Center at Stanford for Cancer Early Detection, Palo Alto, CA, 94304, USA
| | - Mehmet Ozgun Ozen
- Department of Radiology Stanford School of Medicine, BioAcoustic MEMS in Medicine Lab (BAMM), Canary Center at Stanford for Cancer Early Detection, Palo Alto, CA, 94304, USA
| | - Ibrahim Kecoglu
- Department of Physics, Bogazici University, Istanbul, 34342, Turkey
| | - Batuhan Koyuncu
- Department of Computer Engineering, Bogazici University, Istanbul, 34342, Turkey
| | - Hulya Torun
- Koc University Graduate School of Sciences and Engineering, Istanbul, 34450, Turkey
- Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, 34450, Turkey
| | - Davod Khalafkhany
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Apoptosis and Cancer Immunology Laboratory (AKiL), Bogazici University, Istanbul, 34342, Turkey
| | - Irem Loc
- Department of Physics, Bogazici University, Istanbul, 34342, Turkey
| | - Mehmet Giray Ogut
- Department of Radiology Stanford School of Medicine, BioAcoustic MEMS in Medicine Lab (BAMM), Canary Center at Stanford for Cancer Early Detection, Palo Alto, CA, 94304, USA
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, 06800, Turkey
- Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, 06800, Turkey
| | - Demir Akin
- Department of Radiology Stanford School of Medicine, BioAcoustic MEMS in Medicine Lab (BAMM), Canary Center at Stanford for Cancer Early Detection, Palo Alto, CA, 94304, USA
| | - Ihsan Solaroglu
- Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, 34450, Turkey
- School of Medicine, Koc University, Istanbul, 34450, Turkey
| | - Nesrin Ozoren
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Apoptosis and Cancer Immunology Laboratory (AKiL), Bogazici University, Istanbul, 34342, Turkey
| | - Mehmet Burcin Unlu
- Department of Physics, Bogazici University, Istanbul, 34342, Turkey
- Faculty of Engineering, Hokkaido University, North-13 West-8, Kita-ku, Sapporo, Hokkaido, 060-8628, Japan
- Global Center for Biomedical Science and Engineering Quantum Medical Science and Engineering (GI-CoRE Cooperating Hub), Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Utkan Demirci
- Department of Radiology Stanford School of Medicine, BioAcoustic MEMS in Medicine Lab (BAMM), Canary Center at Stanford for Cancer Early Detection, Palo Alto, CA, 94304, USA
| |
Collapse
|
34
|
Chen M, Lin S, Zhou C, Cui D, Haick H, Tang N. From Conventional to Microfluidic: Progress in Extracellular Vesicle Separation and Individual Characterization. Adv Healthc Mater 2023; 12:e2202437. [PMID: 36541411 DOI: 10.1002/adhm.202202437] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles, which contain a wide variety of cargo such as proteins, miRNAs, and lipids. A growing body of evidence suggests that EVs are promising biomarkers for disease diagnosis and therapeutic strategies. Although the excellent clinical value, their use in personalized healthcare practice is not yet feasible due to their highly heterogeneous nature. Taking the difficulty of isolation and the small size of EVs into account, the characterization of EVs at a single-particle level is both imperative and challenging. In a bid to address this critical point, more research has been directed into a microfluidic platform because of its inherent advantages in sensitivity, specificity, and throughput. This review discusses the biogenesis and heterogeneity of EVs and takes a broad view of state-of-the-art advances in microfluidics-based EV research, including not only EV separation, but also the single EV characterization of biophysical detection and biochemical analysis. To highlight the advantages of microfluidic techniques, conventional technologies are included for comparison. The current status of artificial intelligence (AI) for single EV characterization is then presented. Furthermore, the challenges and prospects of microfluidics and its combination with AI applications in single EV characterization are also discussed. In the foreseeable future, recent breakthroughs in microfluidic platforms are expected to pave the way for single EV analysis and improve applications for precision medicine.
Collapse
Affiliation(s)
- Mingrui Chen
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shujing Lin
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Cheng Zhou
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Daxiang Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Hossam Haick
- Department of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ning Tang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
35
|
An N, Bi C, Liu H, Zhao L, Chen X, Chen M, Chen J, Yang S. Shape-Preserving Transformation of Electrodeposited Macroporous Microparticles for Single-Particle SERS Applications. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8286-8297. [PMID: 36719779 DOI: 10.1021/acsami.2c18314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Microparticles composed of bicontinuous and ordered macropores are important in many applications. However, rational integration of ordered macropores into a single crystalline microparticle remains a challenge. Here, we report a method to prepare three-dimensionally ordered macroporous (3DOM) Ag7O8NO3 micropyramids via selectively cementing the colloidal crystal templates via an electrochemical method and their shape-preserving transformation into 3DOM Ag micropryamids formed by Ag nanoparticles via a chemical reduction process. The interconnected macropores facilitated the transportation and enrichment of the analyte molecules into the 3DOM Ag micropyramids. The dense Ag nanoparticles on the skeletons of the 3DOM Ag micropyramids provided strong electromagnetic fields. Taken together, a 3DOM Ag micropyramid as a kind of single-particle surface-enhanced Raman scattering (SERS) sensing substrate demonstrated high SERS sensitivity and outstanding SERS signal reproducibility. We explored the application of 3DOM Ag micropyramids in SERS detection of biomolecules (e.g., adenosine, adenine, hemoglobin bovine, and lysozyme) and proved their potentials in distinguishing exosomes from tumor and non-tumor cells. The method can be extended to prepared 3DOM structures of other materials with promising applications in sensing, separation, and catalytic fields.
Collapse
Affiliation(s)
- Ning An
- School of Materials Science and Engineering, Institute for Composites Science Innovation, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Chao Bi
- Core Facilities, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Hong Liu
- School of Materials Science and Engineering, Institute for Composites Science Innovation, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Liyan Zhao
- School of Materials Science and Engineering, Institute for Composites Science Innovation, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Xueyan Chen
- School of Materials Science and Engineering, Institute for Composites Science Innovation, Zhejiang University, Hangzhou, Zhejiang310027, China
| | - Ming Chen
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Jing Chen
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Shikuan Yang
- School of Materials Science and Engineering, Institute for Composites Science Innovation, Zhejiang University, Hangzhou, Zhejiang310027, China
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, Zhejiang310027, China
| |
Collapse
|
36
|
Jonak ST, Liu Z, Liu J, Li T, D'Souza BV, Schiaffino JA, Oh S, Xie YH. Analyzing bronchoalveolar fluid derived small extracellular vesicles using single-vesicle SERS for non-small cell lung cancer detection. SENSORS & DIAGNOSTICS 2023; 2:90-99. [PMID: 36741247 PMCID: PMC9850358 DOI: 10.1039/d2sd00109h] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
An emerging body of research by biologists and clinicians has demonstrated the clinical application of small extracellular vesicles (sEVs, also commonly referred to as exosomes) as biomarkers for cancer detections. sEVs isolated from various body fluids such as blood, saliva, urine, and cerebrospinal fluid have been used for biomarker discoveries with highly encouraging outcomes. Among the biomarkers discovered are those responsible for multiple cancer types and immune responses. These biomarkers are recapitulated from the tumor microenvironments. Yet, despite numerous discussions of sEVs in scientific literature, sEV-based biomarkers have so far played only a minor role for cancer diagnostics in the clinical setting, notably less so than other techniques such as imaging and biopsy. In this paper, we report the results of a pilot study (n = 10 from each of the patient and the control group) using bronchoalveolar lavage fluid to determine the presence of sEVs related to non-small cell lung cancer in twenty clinical samples examined using surface enhanced Raman spectroscopy (SERS).
Collapse
Affiliation(s)
- Sumita T. Jonak
- NurLabsSan AntonioTX 78201USA,UCLA Biodesign, University of California Los AngelesLos AngelesCA 90095USA
| | - Zirui Liu
- Department of Materials Science and Engineering, University of California Los AngelesLos AngelesCA 90095USA(310) 259 6946
| | - Jun Liu
- Department of Materials Science and Engineering, University of California Los AngelesLos AngelesCA 90095USA(310) 259 6946
| | - Tieyi Li
- Department of Materials Science and Engineering, University of California Los AngelesLos AngelesCA 90095USA(310) 259 6946
| | - Brian V. D'Souza
- NurLabsSan AntonioTX 78201USA,UCLA Biodesign, University of California Los AngelesLos AngelesCA 90095USA
| | - J. Alan Schiaffino
- NurLabsSan AntonioTX 78201USA,UCLA Biodesign, University of California Los AngelesLos AngelesCA 90095USA
| | - Scott Oh
- NurLabsSan AntonioTX 78201USA,UCLA Biodesign, University of California Los AngelesLos AngelesCA 90095USA,UCLA Health System, University of California Los AngelesLos AngelesCA 90095USA
| | - Ya-Hong Xie
- Department of Materials Science and Engineering, University of California Los AngelesLos AngelesCA 90095USA(310) 259 6946,NurLabsSan AntonioTX 78201USA,UCLA Biodesign, University of California Los AngelesLos AngelesCA 90095USA,UCLA Jonsson Comprehensive Cancer Center, University of California, Los AngelesLos AngelesCA 90095USA
| |
Collapse
|
37
|
Jin M, Zhang S, Wang M, Li Q, Ren J, Luo Y, Sun X. Exosomes in pathogenesis, diagnosis, and therapy of ischemic stroke. Front Bioeng Biotechnol 2022; 10:980548. [PMID: 36588958 PMCID: PMC9800834 DOI: 10.3389/fbioe.2022.980548] [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: 06/28/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Ischemic stroke is one of the major contributors to death and disability worldwide. Thus, there is an urgent need to develop early brain tissue perfusion therapies following acute stroke and to enhance functional recovery in stroke survivors. The morbidity, therapy, and recovery processes are highly orchestrated interactions involving the brain with other tissues. Exosomes are natural and ideal mediators of intercellular information transfer and recognized as biomarkers for disease diagnosis and prognosis. Changes in exosome contents express throughout the physiological process. Accumulating evidence demonstrates the use of exosomes in exploring unknown cellular and molecular mechanisms of intercellular communication and organ homeostasis and indicates their potential role in ischemic stroke. Inspired by the unique properties of exosomes, this review focuses on the communication, diagnosis, and therapeutic role of various derived exosomes, and their development and challenges for the treatment of cerebral ischemic stroke.
Collapse
Affiliation(s)
- Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Shuxia Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Mengchen Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Qiaoyu Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Jiahui Ren
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China,*Correspondence: Yun Luo, ; Xiaobo Sun,
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China,*Correspondence: Yun Luo, ; Xiaobo Sun,
| |
Collapse
|
38
|
Fang X, Wang Y, Wang S, Liu B. Nanomaterials assisted exosomes isolation and analysis towards liquid biopsy. Mater Today Bio 2022; 16:100371. [PMID: 35937576 PMCID: PMC9352971 DOI: 10.1016/j.mtbio.2022.100371] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/18/2022] Open
Abstract
Exosomes has attracted tremendous research interests as they are emerging as a new paradigm of liquid biopsy. Although the concentration of exosomes in blood is relatively abundant, there still exists various vesicle-like nanoparticles, such as microvesicles, apoptotic bodies. It's an urgent need to isolate and enrich exosomes from the complex contaminants in biofluid samples. Moreover, the expressing level of exosomal biomarkers varies a lot, which make the sensitive molecular detection of exosomes in high demand. Unfortunately, the efficient isolation and sensitive molecular quantification of exosomes is still a major obstacle hindering the further development and clinical application of exosome-based liquid biopsy. Nanomaterials, with unique physiochemical properties, have been widely used in biosensing and analysis aspects, thus they are thought as powerful tools for effective purification and molecular analysis of exosomes. In this review, we summarized the most recent progresses in nanomaterials assisted exosome isolation and analysis towards liquid biopsy. On the one hand, nanomaterials can be used as capture substrates to afford large binding area and specific affinity to exosomes. Meanwhile, nanomaterials can also be served as promising signal transducers and amplifiers for molecular detection of exosomes. Furthermore, we also pointed out several potential and promising research directions in nanomaterials assisted exosome analysis. It's envisioned that this review will give the audience a complete outline of nanomaterials in exosome study, and further promote the intersection of nanotechnology and bio-analysis.
Collapse
Affiliation(s)
- Xiaoni Fang
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Yuqing Wang
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Shurong Wang
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Baohong Liu
- School of Pharmacy, Shanghai Stomatological Hospital, Department of Chemistry, Fudan University, Shanghai, 200438, China
| |
Collapse
|
39
|
Zheng H, Ding Q, Li C, Chen W, Chen X, Lin Q, Wang D, Weng Y, Lin D. Recent progress in surface-enhanced Raman spectroscopy-based biosensors for the detection of extracellular vesicles. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:4161-4173. [PMID: 36254847 DOI: 10.1039/d2ay01339h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Extracellular vesicles (EVs) are a type of mediator that enables intercellular communication. Moreover, EVs carry critical molecular information from parental cells, making them ideal biomarkers for clinical screening and diagnosis. Currently, several sensing technologies have been established to sensitively detect EVs. Among them, surface-enhanced Raman spectroscopy (SERS) has become a powerful analytical tool with high sensitivity and low detection limits. In this review, we first cover the biological characteristics of EVs and the principle of SERS amplification. Then, we describe the recent progress in SERS technology applied to detect EVs, including direct label-free methods and indirect labeling strategies, in which substrate fabrication and nanoprobe assembly were emphasized. Furthermore, SERS technology could also be used to characterize or monitor the behavior of programmable EVs. Finally, we discuss the prospects and issues to be addressed for the development of SERS technology for EV analysis.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Qin Ding
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Chen Li
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Wei Chen
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Xiaoqiang Chen
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Qin Lin
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Desheng Wang
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Youliang Weng
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Duo Lin
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, China
| |
Collapse
|
40
|
Tavakkoli Yaraki M, Tukova A, Wang Y. Emerging SERS biosensors for the analysis of cells and extracellular vesicles. NANOSCALE 2022; 14:15242-15268. [PMID: 36218172 DOI: 10.1039/d2nr03005e] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cells and their derived extracellular vesicles (EVs) or exosomes contain unique molecular signatures that could be used as biomarkers for the detection of severe diseases such as cancer, as well as monitoring the treatment response. Revealing these molecular signatures requires developing non-invasive ultrasensitive tools to enable single molecule/cell-level detection using a small volume of sample with low signal-to-noise ratio background and multiplex capability. Surface-enhanced Raman scattering (SERS) can address the current limitations in studying cells and EVs through two main mechanisms: plasmon-enhanced electric field (the so-called electromagnetic mechanism (EM)), and chemical mechanism (CM). In this review, we first highlight these two SERS mechanisms and then discuss the nanomaterials that have been used to develop SERS biosensors based on each of the aforementioned mechanisms as well as the combination of these two mechanisms in order to take advantage of the synergic effect between electromagnetic enhancement and chemical enhancement. Then, we review the recent advances in designing label-aided and label-free SERS biosensors in both colloidal and planar systems to investigate the surface biomarkers on cancer cells and their derived EVs. Finally, we discuss perspectives of emerging SERS biosensors in future biomedical applications. We believe this review article will thus appeal to researchers in the field of nanobiotechnology including material sciences, biosensors, and biomedical fields.
Collapse
Affiliation(s)
- Mohammad Tavakkoli Yaraki
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| | - Anastasiia Tukova
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
41
|
Faur CI, Roman RC, Jurj A, Raduly L, Almășan O, Rotaru H, Chirilă M, Moldovan MA, Hedeșiu M, Dinu C. Salivary Exosomal MicroRNA-486-5p and MicroRNA-10b-5p in Oral and Oropharyngeal Squamous Cell Carcinoma. Medicina (B Aires) 2022; 58:medicina58101478. [PMID: 36295638 PMCID: PMC9610161 DOI: 10.3390/medicina58101478] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
Background and Objectives: The research aimed at evaluating the capacity of salivary exosomal miR-10b-5p and miR-486-5p for oral and oropharyngeal cancer detection. Materials and Methods: The saliva samples were harvested from histopathological diagnosed oral and oropharyngeal squamous cell carcinoma patients and healthy volunteer subjects. The exosomes were isolated by differential ultracentrifugation and quantified by Nano Track Analysis. The microRNAs were extracted and quantified from salivary exosomes by quantitative Real-Time Polymerase Chain Reaction. Results: This research comprised fifty participants. When compared to healthy controls, salivary exosomal miR-486-5p was elevated and miR-10b-5p was reduced in oral and oropharyngeal squamous cell carcinoma. Moreover, miR-486-5p had a high expression level in stage II of cancer in comparison to the other cancer stages. The cancer samples presented an increased exosome dimension compared to the control samples. Conclusions: Salivary exosomal miR-10b-5p and miR-486-5p have an altered expression in oral and oropharyngeal cancer.
Collapse
Affiliation(s)
- Cosmin Ioan Faur
- Department of Maxillofacial Surgery and Radiology, Oral Radiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 32 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Rareș Călin Roman
- Department of Maxillofacial Surgery and Radiology, Oral and Cranio-Maxillofacial Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, 33 Moților Street, 400001 Cluj-Napoca, Romania
- Correspondence:
| | - Ancuța Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Oana Almășan
- Department of Prosthetic Dentistry and Dental Materials, “Iuliu Hațieganu” University of Medicine and Pharmacy, 32 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Horațiu Rotaru
- Department of Maxillofacial Surgery and Radiology, Oral and Cranio-Maxillofacial Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, 33 Moților Street, 400001 Cluj-Napoca, Romania
| | - Magdalena Chirilă
- 8th Department-Surgical Secialties, O.R.L., “Iuliu Hațieganu” University of Medicine and Pharmacy, 4-6 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Mădălina Anca Moldovan
- Department of Maxillofacial Surgery and Radiology, Oral and Cranio-Maxillofacial Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, 33 Moților Street, 400001 Cluj-Napoca, Romania
| | - Mihaela Hedeșiu
- Department of Maxillofacial Surgery and Radiology, Oral Radiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 32 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Cristian Dinu
- Department of Maxillofacial Surgery and Radiology, Maxillofacial Surgery and Implantology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 37 Iuliu Hossu Street, 400429 Cluj-Napoca, Romania
| |
Collapse
|
42
|
Wang Y, Zhu J, Chen J, Xu R, Groth T, Wan H, Zhou G. The Signaling Pathways Induced by Exosomes in Promoting Diabetic Wound Healing: A Mini-Review. Curr Issues Mol Biol 2022; 44:4960-4976. [PMID: 36286052 PMCID: PMC9600352 DOI: 10.3390/cimb44100337] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Impaired healing of diabetic wounds harms patients' quality of life and even leads to disability and death, which is an urgent issue to be solved clinically. Despite the great progress that has been achieved, it remains a worldwide challenge to develop effective therapeutic treatments for diabetic wounds. Recently, exosomes have attracted special attention because they can be involved in immune response, antigen presentation, cell migration, cell differentiation, tumor invasion and other processes. Meanwhile, exosomes have been proven to hold great potential in the treatment of diabetic wounds. Mechanistic studies of exosomes based on signaling pathways could not only help to uncover the mechanisms by which exosomes promote diabetic wound healing but could also provide a theoretical basis for the clinical application of exosomes. Herein, our mini-review aims to summarize the progress of research on the use of various exosomes derived from different cell types to promote diabetic wound healing, with a focus on the classical signaling pathways, including PI3K/Akt, Wnt, NF-κB, MAPK, Notch, Nrf2, HIF-1α/VEGF and TGF-β/Smad. The results show that exosomes could regulate these signaling pathways to down-regulate inflammation, reduce oxidative stress, increase angiogenesis, promote fibroblast proliferation, induce re-epithelization and inhibit scar formation, making exosomes attractive candidates for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Yanying Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Jiayan Zhu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Jing Chen
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Ruojiao Xu
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, D-06099 Halle (Saale), Germany
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- Correspondence: (H.W.); (G.Z.)
| | - Guoying Zhou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- College of Life Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
- Correspondence: (H.W.); (G.Z.)
| |
Collapse
|
43
|
Xie Y, Su X, Wen Y, Zheng C, Li M. Artificial Intelligent Label-Free SERS Profiling of Serum Exosomes for Breast Cancer Diagnosis and Postoperative Assessment. NANO LETTERS 2022; 22:7910-7918. [PMID: 36149810 DOI: 10.1021/acs.nanolett.2c02928] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Breast cancer subtypes have important implications of treatment responses and clinical outcomes. Exosomes have been considered as promising biomarkers for liquid biopsies, but the utility of exosomes for accurate diagnosis of distinct breast cancer subtypes is a grand challenge due to the difficulty in uncovering the subtle compositional difference in complex clinical settings. Herein, we report an artificial intelligent surface-enhanced Raman spectroscopy (SERS) strategy for label-free spectroscopic analysis of serum exosomes, allowing for accurate diagnosis of breast cancer and assessment of surgical outcomes. Our deep learning algorithm trained with SERS spectra of cancer cell-derived exosomes is demonstrated with a 100% prediction accuracy for human patients with different breast cancer subtypes who do not undergo surgery using SERS spectra of serum exosomes. Furthermore, when combined with similarity analysis by principal component analysis, our approach is able to evaluate the surgical outcomes of breast cancer of distinct molecular subtypes.
Collapse
Affiliation(s)
- Yangcenzi Xie
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Xiaoming Su
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, China
| | - Yu Wen
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Chao Zheng
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
44
|
Leggio L, L'Episcopo F, Magrì A, Ulloa‐Navas MJ, Paternò G, Vivarelli S, Bastos CAP, Tirolo C, Testa N, Caniglia S, Risiglione P, Pappalardo F, Serra A, García‐Tárraga P, Faria N, Powell JJ, Peruzzotti‐Jametti L, Pluchino S, García‐Verdugo JM, Messina A, Marchetti B, Iraci N. Small Extracellular Vesicles Secreted by Nigrostriatal Astrocytes Rescue Cell Death and Preserve Mitochondrial Function in Parkinson's Disease. Adv Healthc Mater 2022; 11:e2201203. [PMID: 35856921 PMCID: PMC11468249 DOI: 10.1002/adhm.202201203] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/05/2022] [Indexed: 01/28/2023]
Abstract
Extracellular vesicles (EVs) are emerging as powerful players in cell-to-cell communication both in healthy and diseased brain. In Parkinson's disease (PD)-characterized by selective dopaminergic neuron death in ventral midbrain (VMB) and degeneration of their terminals in striatum (STR)-astrocytes exert dual harmful/protective functions, with mechanisms not fully elucidated. Here, this study shows that astrocytes from the VMB-, STR-, and VMB/STR-depleted brains release a population of small EVs in a region-specific manner. Interestingly, VMB-astrocytes secreted the highest rate of EVs, which is further exclusively increased in response to CCL3, a chemokine that promotes robust dopaminergic neuroprotection in different PD models. The neuroprotective potential of nigrostriatal astrocyte-EVs is investigated in differentiated versus undifferentiated SH-SY5Y cells exposed to oxidative stress and mitochondrial toxicity. EVs from both VMB- and STR-astrocytes counteract H2 O2 -induced caspase-3 activation specifically in differentiated cells, with EVs from CCL3-treated astrocytes showing a higher protective effect. High resolution respirometry further reveals that nigrostriatal astrocyte-EVs rescue neuronal mitochondrial complex I function impaired by the neurotoxin MPP+ . Notably, only EVs from VMB-astrocyte fully restore ATP production, again specifically in differentiated SH-SY5Y. These results highlight a regional diversity in the nigrostriatal system for the secretion and activities of astrocyte-EVs, with neuroprotective implications for PD.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | - Andrea Magrì
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - María José Ulloa‐Navas
- Laboratory of Compared NeurobiologyUniversity of Valencia‐CIBERNEDPaterna46980Spain
- Department of NeuroscienceMayo ClinicJacksonvilleFL32257USA
| | - Greta Paternò
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | | | | | | | - Pierpaolo Risiglione
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - Fabrizio Pappalardo
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | | | - Nuno Faria
- Department of Veterinary MedicineUniversity of CambridgeCambridgeCB3 0ESUK
| | - Jonathan J. Powell
- Department of Veterinary MedicineUniversity of CambridgeCambridgeCB3 0ESUK
| | | | - Stefano Pluchino
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | | | - Angela Messina
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
- Oasi Research Institute‐IRCCSTroina94018Italy
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| |
Collapse
|
45
|
Liu Z, Li T, Wang Z, Liu J, Huang S, Min BH, An JY, Kim KM, Kim S, Chen Y, Liu H, Kim Y, Wong DT, Huang TJ, Xie YH. Gold Nanopyramid Arrays for Non-Invasive Surface-Enhanced Raman Spectroscopy-Based Gastric Cancer Detection via sEVs. ACS APPLIED NANO MATERIALS 2022; 5:12506-12517. [PMID: 36185166 PMCID: PMC9513748 DOI: 10.1021/acsanm.2c01986] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/12/2022] [Indexed: 05/05/2023]
Abstract
Gastric cancer (GC) is one of the most common and lethal types of cancer affecting over one million people, leading to 768,793 deaths globally in 2020 alone. The key for improving the survival rate lies in reliable screening and early diagnosis. Existing techniques including barium-meal gastric photofluorography and upper endoscopy can be costly and time-consuming and are thus impractical for population screening. We look instead for small extracellular vesicles (sEVs, currently also referred as exosomes) sized ⌀ 30-150 nm as a candidate. sEVs have attracted a significantly higher level of attention during the past decade or two because of their potentials in disease diagnoses and therapeutics. Here, we report that the composition information of the collective Raman-active bonds inside sEVs of human donors obtained by surface-enhanced Raman spectroscopy (SERS) holds the potential for non-invasive GC detection. SERS was triggered by the substrate of gold nanopyramid arrays we developed previously. A machine learning-based spectral feature analysis algorithm was developed for objectively distinguishing the cancer-derived sEVs from those of the non-cancer sub-population. sEVs from the tissue, blood, and saliva of GC patients and non-GC participants were collected (n = 15 each) and analyzed. The algorithm prediction accuracies were reportedly 90, 85, and 72%. "Leave-a-pair-of-samples out" validation was further performed to test the clinical potential. The area under the curve of each receiver operating characteristic curve was 0.96, 0.91, and 0.65 in tissue, blood, and saliva, respectively. In addition, by comparing the SERS fingerprints of individual vesicles, we provided a possible way of tracing the biogenesis pathways of patient-specific sEVs from tissue to blood to saliva. The methodology involved in this study is expected to be amenable for non-invasive detection of diseases other than GC.
Collapse
Affiliation(s)
- Zirui Liu
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Tieyi Li
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Zeyu Wang
- Department
of Mechanical Engineering and Material Science, Duke University, Durham, North Carolina 27708, United States
| | - Jun Liu
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Shan Huang
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Byoung Hoon Min
- Department
of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Ji Young An
- Department
of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Kyoung Mee Kim
- Department
of Pathology and Translational Genomics, Sungkyunkwan University School
of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Sung Kim
- Department
of Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 135-710, Korea
| | - Yiqing Chen
- Department
of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| | - Huinan Liu
- Department
of Bioengineering, University of California, Riverside, Riverside, California 92521, United States
| | - Yong Kim
- UCLA
School of Dentistry, 10833 Le Conte Ave. Box 951668, Los Angeles, California 90095-1668, United States
| | - David T.W. Wong
- UCLA
School of Dentistry, 10833 Le Conte Ave. Box 951668, Los Angeles, California 90095-1668, United States
| | - Tony Jun Huang
- Department
of Mechanical Engineering and Material Science, Duke University, Durham, North Carolina 27708, United States
| | - Ya-Hong Xie
- Department
of Materials Science and Engineering, University
of California Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
46
|
Hamzah R, Alghazali K, Biris A, Griffin RJ. Nanoparticle-Labeled Exosomes as Theranostic Agents: A Review. ACS APPLIED NANO MATERIALS 2022; 5:12265-12275. [DOI: 10.1021/acsanm.2c01426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Rabab Hamzah
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Karrer Alghazali
- Nushores Biosciences LLC, Little Rock, Arkansas 72211, United States
| | - Alexandru Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas 72204, United States
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
47
|
Dutta S, Nimse SB, Van Keuren-Jensen K, Bitan G. Editorial: Exosomes: Message in a vesicle. Front Pharmacol 2022; 13:1018928. [PMID: 36160457 PMCID: PMC9494591 DOI: 10.3389/fphar.2022.1018928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Suman Dutta
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Suman Dutta,
| | - Satish Balasaheb Nimse
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon, South Korea
| | | | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
48
|
Al Mannai A, Al-Ansari T, Saoud KM. Quantification of Serum Exosome Biomarkers Using 3D Nanoporous Gold and Spectrophotometry. SENSORS (BASEL, SWITZERLAND) 2022; 22:6347. [PMID: 36080806 PMCID: PMC9460504 DOI: 10.3390/s22176347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 06/15/2023]
Abstract
Tumor-derived exosomes may provide biomarkers for cancer treatment. Using sputtering technology, an affinity-based device to capture exosomes was developed using nanoporous substrate (NPG)-coated silicon microscopy. Immunology-based techniques detect and purify exosomes using gold coating with a specific antigen. Inverted fluorescent microscopy was used to detect target exosomes quantitatively utilizing fluorescent nanospheres as the label. We quantified the expression of CD63 surface protein markers on exosomes from conditioned culture media of breast cancer cells. The exosomes that targeted specific proteins with controls were statistically analyzed and compared to those that targeted non-specific proteins. Results from SEM showed that the exosomes were circular, between 30 and 150 nanometers in size. The porous gold substrates captured more exosomes than the nonporous substrates. Nitric acid treatments at different times resulted in a variety of pore sizes. Despite the increase in the size of the pores, the number of exosomes found in the porous gold substrate treated for 10 min nearly doubled compared to the one treated for 5 min. In this work, a fluorescence biosensor was developed to detect breast cancer exosomes using nanoporous gold substrates (NPG). Assay and model exosomes of specific breast cancer cells showed that exosomes exhibit diagnostic surface protein markers, reflecting the protein profile of their parent cells. Furthermore, the specific binding between the exosome surface antibodies and the targets identified the CD63 biomarkers on the exosome, suggesting these markers' diagnostic potential. This study can accelerate exosome research in determining tumor-related exosomes and develop novel cancer diagnostic methods.
Collapse
Affiliation(s)
- Amera Al Mannai
- College of Science and Engineering, Hamad Bin Khalifa University, Doha P.O. Box 34111, Qatar
| | - Tareq Al-Ansari
- College of Science and Engineering, Hamad Bin Khalifa University, Doha P.O. Box 34111, Qatar
| | - Khaled M. Saoud
- Liberal Arts and Science Program, Virginia Commonwealth University, Doha P.O. Box 8095, Qatar
| |
Collapse
|
49
|
Mateescu B, Jones JC, Alexander RP, Alsop E, An JY, Asghari M, Boomgarden A, Bouchareychas L, Cayota A, Chang HC, Charest A, Chiu DT, Coffey RJ, Das S, De Hoff P, deMello A, D’Souza-Schorey C, Elashoff D, Eliato KR, Franklin JL, Galas DJ, Gerstein MB, Ghiran IH, Go DB, Gould S, Grogan TR, Higginbotham JN, Hladik F, Huang TJ, Huo X, Hutchins E, Jeppesen DK, Jovanovic-Talisman T, Kim BY, Kim S, Kim KM, Kim Y, Kitchen RR, Knouse V, LaPlante EL, Lebrilla CB, Lee LJ, Lennon KM, Li G, Li F, Li T, Liu T, Liu Z, Maddox AL, McCarthy K, Meechoovet B, Maniya N, Meng Y, Milosavljevic A, Min BH, Morey A, Ng M, Nolan J, De Oliveira Junior GP, Paulaitis ME, Phu TA, Raffai RL, Reátegui E, Roth ME, Routenberg DA, Rozowsky J, Rufo J, Senapati S, Shachar S, Sharma H, Sood AK, Stavrakis S, Stürchler A, Tewari M, Tosar JP, Tucker-Schwartz AK, Turchinovich A, Valkov N, Van Keuren-Jensen K, Vickers KC, Vojtech L, Vreeland WN, Wang C, Wang K, Wang Z, Welsh JA, Witwer KW, Wong DT, Xia J, Xie YH, Yang K, Zaborowski MP, Zhang C, Zhang Q, Zivkovic AM, Laurent LC. Phase 2 of extracellular RNA communication consortium charts next-generation approaches for extracellular RNA research. iScience 2022; 25:104653. [PMID: 35958027 PMCID: PMC9358052 DOI: 10.1016/j.isci.2022.104653] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The extracellular RNA communication consortium (ERCC) is an NIH-funded program aiming to promote the development of new technologies, resources, and knowledge about exRNAs and their carriers. After Phase 1 (2013-2018), Phase 2 of the program (ERCC2, 2019-2023) aims to fill critical gaps in knowledge and technology to enable rigorous and reproducible methods for separation and characterization of both bulk populations of exRNA carriers and single EVs. ERCC2 investigators are also developing new bioinformatic pipelines to promote data integration through the exRNA atlas database. ERCC2 has established several Working Groups (Resource Sharing, Reagent Development, Data Analysis and Coordination, Technology Development, nomenclature, and Scientific Outreach) to promote collaboration between ERCC2 members and the broader scientific community. We expect that ERCC2's current and future achievements will significantly improve our understanding of exRNA biology and the development of accurate and efficient exRNA-based diagnostic, prognostic, and theranostic biomarker assays.
Collapse
Affiliation(s)
- Bogdan Mateescu
- Brain Research Institute, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Jennifer C. Jones
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric Alsop
- Neurogenomics Division, TGen, Phoenix, AZ 85004, USA
| | - Ji Yeong An
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Mohammad Asghari
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Alex Boomgarden
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Laura Bouchareychas
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Alfonso Cayota
- Functional Genomics Unit, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- University Hospital, Universidad de la República, Montevideo 11600, Uruguay
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Al Charest
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel T. Chiu
- Department of Chemistry and Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Robert J. Coffey
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Peter De Hoff
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, La Jolla, San Diego, CA 92093, USA
| | - Andrew deMello
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | | | - David Elashoff
- Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Kiarash R. Eliato
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Jeffrey L. Franklin
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - David J. Galas
- Pacific Northwest Research Institute, Seattle, WA 98122, USA
| | - Mark B. Gerstein
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT 06520, USA
- Department of Computer Science, Yale University, New Haven, CT 06520, USA
| | - Ionita H. Ghiran
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David B. Go
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Stephen Gould
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Tristan R. Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA, USA
| | - James N. Higginbotham
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Florian Hladik
- Departments of Obstetrics and Gynecology, and Medicine, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tony Jun Huang
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Xiaoye Huo
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Dennis K. Jeppesen
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tijana Jovanovic-Talisman
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Betty Y.S. Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sung Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyoung-Mee Kim
- Department of Pathology & Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yong Kim
- Department of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Robert R. Kitchen
- Corrigan Minehan Heart Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vaughan Knouse
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Emily L. LaPlante
- Bioinformatics Research Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - L. James Lee
- Department of Chemical and Biomolecular Engineering and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Feng Li
- Department of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Tieyi Li
- Department of Materials Science & Engineering, University of California Los Angeles, Los Angeles, CA 90095-1595, USA
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Zirui Liu
- Department of Materials Science & Engineering, University of California Los Angeles, Los Angeles, CA 90095-1595, USA
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Kyle McCarthy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Nalin Maniya
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Yingchao Meng
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Aleksandar Milosavljevic
- Bioinformatics Research Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Program in Quantitative and Computational Biosciences Baylor College of Medicine, Houston, TX 77030, USA
| | - Byoung-Hoon Min
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, South Korea
| | - Amber Morey
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, La Jolla, San Diego, CA 92093, USA
| | - Martin Ng
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - John Nolan
- Scintillon Institute, San Diego, CA, USA
| | | | - Michael E. Paulaitis
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tuan Anh Phu
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Robert L. Raffai
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Northern California Institute for Research and Education, San Francisco, CA 94121, USA
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA
| | - Eduardo Reátegui
- Department of Chemical and Biomolecular Engineering and Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Matthew E. Roth
- Bioinformatics Research Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Joel Rozowsky
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Joseph Rufo
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sigal Shachar
- Meso Scale Diagnostics, LLC, Rockville, MD 20850, USA
| | - Himani Sharma
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology & Reproductive Medicine, University of Texas MD Aderson Cancer Center, Houston, TX 77030, USA
| | - Stavros Stavrakis
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Alessandra Stürchler
- Brain Research Institute, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir Prelog Weg 1, 8093 Zürich, Switzerland
| | - Muneesh Tewari
- Department of Internal Medicine, Hematology/Oncology Division, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Juan P. Tosar
- Functional Genomics Unit, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay
| | | | - Andrey Turchinovich
- Cancer Genome Research (B063), German Cancer Research Center DKFZ, Heidelberg 69120, Germany
- Heidelberg Biolabs GmbH, Heidelberg 69120, Germany
| | - Nedyalka Valkov
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Kasey C. Vickers
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Wyatt N. Vreeland
- Bioprocess Measurement Group, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Ceming Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - ZeYu Wang
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Joshua A. Welsh
- Laboratory of Pathology Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David T.W. Wong
- Department of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Jianping Xia
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Ya-Hong Xie
- Department of Materials Science & Engineering, University of California Los Angeles, Los Angeles, CA 90095-1595, USA
| | - Kaichun Yang
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC 27708, USA
| | - Mikołaj P. Zaborowski
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznań, Poland
| | - Chenguang Zhang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Qin Zhang
- Department of Medicine/Gastroenterology and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Louise C. Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, La Jolla, San Diego, CA 92093, USA
| |
Collapse
|
50
|
Abstract
Exosomes are a type of extracellular vesicles secreted by cells in normal or pathological conditions for cell-cell communication. With immunomodulatory characteristics and potential therapeutic properties, immune-cell-derived exosomes play an important role in cancer therapy. They express various antigens on their surface, which can be employed for antigen presentation, immunological activation, and metabolic regulation, leading to the killing of cancerous cells. In addition, immune-cell-derived exosomes have received extensive attention as a drug delivery platform in effective antitumor therapy due to their excellent biocompatibility, low immunogenicity, and high loading capacity. In this review, the biological and therapeutic characteristics of immune-cell-derived exosomes are comprehensively outlined. The antitumor mechanism of exosomes secreted by immune cells, including macrophages, dendritic cells, T cells, B cells, and natural killer cells, are systematically summarized. Moreover, the applications of immune-cell-derived exosomes as nanocarriers to transport antitumor agents (chemotherapeutic drugs, genes, proteins, etc.) are discussed. More importantly, the existing challenges of immune-cell-derived exosomes are pointed out, and their antitumor potentials are also discussed.
Collapse
Affiliation(s)
- Yongmei Zhao
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales 2145, Australia
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong 226019, China
| |
Collapse
|