1
|
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
|
2
|
Zhang G, Huang X, Gong Y, Ding Y, Wang H, Zhang H, Wu L, Su R, Yang C, Zhu Z. Fingerprint Profiling of Glycans on Extracellular Vesicles via Lectin-Induced Aggregation Strategy for Precise Cancer Diagnostics. J Am Chem Soc 2024; 146:29053-29063. [PMID: 39235449 DOI: 10.1021/jacs.4c10390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Extracellular vesicles (EVs) harbor abundant glycans that mediate various functions, such as intercellular communication and disease advancement, which play significant roles in disease progression. However, the presence of EV heterogeneity in body fluids and the complex nature of the glycan structures have posed challenges for the detection of EV glycans. In this study, we provide a streamlined method integrated, membrane-specific separation with lectin-induced aggregation strategy (MESSAGE), for multiplexed profiling of EV glycans. By leveraging a rationally designed lectin-induced aggregation strategy, the expression of EV glycans is converted to size-based signals. With the assistance learning machine algorithms, the MESSAGE strategy with high sensitivity, specificity, and simplicity can be used for early cancer diagnosis and classification, as well as monitoring cancer metastasis via 20 μL plasma sample within 2 h. Furthermore, our platform holds promise for advancing the field of EV-based liquid biopsy for clinical applications, opening new possibilities for the profiling of EV glycan signatures in various disease states.
Collapse
Affiliation(s)
- Guihua Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaodan Huang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yanli Gong
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yue Ding
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hua Wang
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Huimin Zhang
- Innovation Laboratory for Sciences, Technologies of Energy Materials of Fujian Province, Xiamen 361000, China
| | - Lingling Wu
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Rui Su
- Department of Hematology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Chaoyong Yang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhi Zhu
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
3
|
He K, Cheng Z, Zhang X, Qian Z, Chen J, Li B, Meng F, Yu S, Tang K, Wu YX. Activating Two-Photon Silica Nanoamplifier-Based CHA and FRET for Accurate Ratiometric Bioimaging of Intracellular MicroRNA. Anal Chem 2024; 96:16338-16345. [PMID: 39359231 DOI: 10.1021/acs.analchem.4c03630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
In situ visualization of microRNA (miRNA) in cancer cells and diseased tissues is essential for advancing our comprehension of the onset and progression of associated diseases. Two-photon (TP) imaging, as an imaging technology with high spatiotemporal resolution, deep tissue penetration, and accurate target quantification, has distinctive advantages over single-photon imaging and has attracted increasing attention. Extensive research has been conducted on two-photon dye-doped silica nanoparticles, which exhibit a large two-photon absorption (TPA) cross-section, high fluorescence quantum yield, and excellent biocompatibility. However, the low abundance of RNA in tumor cells leads to insufficient signal output. Based on functional nucleic acid, a catalyzed hairpin self-assembly (CHA) signal amplification strategy, which has simplicity, robustness, and nonenzymatic characteristics, can achieve the amplification of DNA or RNA signals. Here, a two-photon silica nanoamplifier (TP-SNA) utilizing TP dye-doped silica nanoparticles (SiNPs) and functional nucleic acid was constructed, employing triggering catalyzed hairpin self-assembly and fluorescence resonance energy transfer (FRET) for highly sensitive detection and precise TP imaging of endogenous miRNAs in tumor cells and tissues at varying depths. The TP-SNA demonstrated the capability to detect miR-203 with a detection limit of 33 pM. The maximum two-photon tissue penetration depth of the two-photon nanoamplifier was 210 μm. The two-photon nanoamplifier developed in this study makes full use of the advantages of accurate TP ratiometric bioimaging and the CHA signal amplification strategy, which shows good application value for future transformation into clinical diagnosis.
Collapse
Affiliation(s)
- Kangdi He
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Zhen Cheng
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xianmiao Zhang
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Zhiling Qian
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jia Chen
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Bingqian Li
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Fayan Meng
- Chemistry & Physics Department, Frostburg State University, 101 Braddock Rd, Frostburg, Maryland 21532, United States
| | - Shengrong Yu
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, Zhejiang 315211, China
| | - Keqi Tang
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, Zhejiang 315211, China
| | - Yong-Xiang Wu
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
- Ningbo Zhenhai Institute of Mass Spectrometry, Ningbo, Zhejiang 315211, China
| |
Collapse
|
4
|
Deng J, Liu C, Sun J. DNA-Based Nanomaterials for Analysis of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303092. [PMID: 38016069 DOI: 10.1002/adma.202303092] [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: 04/03/2023] [Revised: 07/21/2023] [Indexed: 11/30/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanovesicles comprising a myriad of molecular cargo such as proteins and nucleic acids, playing essential roles in intercellular communication and physiological and pathological processes. EVs have received substantial attention as noninvasive biomarkers for disease diagnosis and prognosis. Owing to their ability to recognize protein and nucleic acid targets, DNA-based nanomaterials with excellent programmability and modifiability provide a promising tool for the sensitive and accurate detection of molecular cargo carried by EVs. In this perspective, recent advancements in EV analysis using a variety of DNA-based nanomaterials are summarized, which can be broadly classified into three categories: linear DNA probes, DNA nanostructures, and hybrid DNA nanomaterials. The design, construction, advantages, and disadvantages of different types of DNA nanomaterials, as well as their performance for detecting EVs are reviewed. The challenges and opportunities in the field of EV analysis by DNA nanomaterials are also discussed.
Collapse
Affiliation(s)
- Jinqi Deng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Liu
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiashu Sun
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
5
|
Sun S, Yang H, Wu Z, Zhang S, Xu J, Shi P. CRISPR/Cas systems combined with DNA nanostructures for biomedical applications. Chem Commun (Camb) 2024; 60:3098-3117. [PMID: 38406926 DOI: 10.1039/d4cc00290c] [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: 02/27/2024]
Abstract
DNA nanostructures are easy to design and construct, have good biocompatibility, and show great potential in biosensing and drug delivery. Numerous distinctive and versatile DNA nanostructures have been developed and explored for biomedical applications. In addition to DNA nanostructures that are completely assembled from DNA, composite DNA nanostructures obtained by combining DNA with other organic or inorganic materials are also widely used in related research. The CRISPR/Cas system has attracted great attention as a powerful gene editing technology and is also widely used in biomedical diagnosis. Many researchers are committed to exploring new possibilities by combining DNA nanostructures with CRISPR/Cas systems. These explorations provide support for the development of new detection methods and cargo delivery pathways, provide inspiration for improving relevant gene editing platforms, and further expand the application scope of DNA nanostructures and CRISPR/Cas systems. This paper mainly reviews the design principles and biomedical applications of CRISPR/Cas combined with DNA nanostructures based on the types of DNA nanostructures. Finally, the application status, challenges and development prospects of CRISPR/Cas combined with DNA nanostructures in detection and delivery are summarized. It is expected that this review will enable researchers to better understand the current state of the field and provide insights into the application of CRISPR/Cas systems and the development of DNA nanostructures.
Collapse
Affiliation(s)
- Shujuan Sun
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi 276000, P. R. China.
| | - Haoqi Yang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi 276000, P. R. China.
| | - Ziyong Wu
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi 276000, P. R. China.
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi 276000, P. R. China.
| | - Jingjuan Xu
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi 276000, P. R. China.
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, P. R. China.
| | - Pengfei Shi
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi 276000, P. R. China.
| |
Collapse
|
6
|
Yang L, Guo H, Gao Q, Hou T, Zhang J, Liu X, Li F. Integrating Reliable Pt-S Bond-Mediated 3D DNA Nanomachine with Magnetic Separation in a Homogeneous Electrochemical Strategy for Exosomal MicroRNA Detection with Low Background and High Sensitivity. Anal Chem 2023; 95:17834-17842. [PMID: 37988125 DOI: 10.1021/acs.analchem.3c03914] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Precise and sensitive analysis of exosomal microRNA (miRNA) is of great importance for noninvasive early disease diagnosis, but it remains a great challenge to detect exosomal miRNA in human blood samples because of their small size, high sequence homology, and low abundance. Herein, we integrated reliable Pt-S bond-mediated three-dimensional (3D) DNA nanomachine and magnetic separation in a homogeneous electrochemical strategy for the detection of exosomal miRNA with low background and high sensitivity. The 3D DNA nanomachine was easily prepared via a facile and rapid freezing method, and it was capable of resisting the influence of biothiols, thus endowing it with high stability. Notably, the as-developed magnetic 3D DNA nanomachine not only enabled the detection system to have a low background but also coupled with liposome nanocarriers to synergistically amplify the current signal. Consequently, by ingeniously combining the low background and multiple signal-amplification strategies in homogeneous electrochemical biosensing, highly sensitive detection of exosomal miRNA was successfully achieved. More significantly, with good anti-interference ability, the as-proposed method could effectively discriminate plasma samples from cancer patients and healthy subjects, thus showing a high potential for application in the nondestructive early clinical diagnosis of disease.
Collapse
Affiliation(s)
- Limin Yang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Heng Guo
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Qian Gao
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Ting Hou
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Jingang Zhang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Xiaojuan Liu
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Feng Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| |
Collapse
|
7
|
Ji S, Wang X, Wang Y, Sun Y, Su Y, Lv X, Song X. Advances in Cas12a-Based Amplification-Free Nucleic Acid Detection. CRISPR J 2023; 6:405-418. [PMID: 37751223 DOI: 10.1089/crispr.2023.0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
In biomedicine, rapid and sensitive nucleic acid detection technology plays an important role in the early detection of infectious diseases. However, most traditional nucleic acid detection methods require the amplification of nucleic acids, resulting in problems such as long detection time, complex operation, and false-positive results. In recent years, clustered regularly interspaced short palindromic repeats (CRISPR) systems have been widely used in nucleic acid detection, especially the CRISPR-Cas12a system, which can trans cleave single-stranded DNA and can realize the detection of DNA targets. But, amplification of nucleic acids is still required to further improve detection sensitivity, which makes Cas12a-based amplification-free nucleic acid detection methods a great challenge. This article reviews the recent progress of Cas12a-based amplification-free detection methods for nucleic acids. These detection methods apply electrochemical detection methods, fluorescence detection methods, noble metal nanomaterial detection methods, and lateral flow assay. Under various optimization strategies, unamplified nucleic acids have the same sensitivity as amplified nucleic acids. At the same time, the article discusses the advantages and disadvantages of each method and further discusses the current challenges such as off-target effects and the ability to achieve high-throughput detection. Amplification-free nucleic acid detection technology based on CRISPR-Cas12a has great potential in the biomedical field.
Collapse
Affiliation(s)
- Shixin Ji
- School of Life Sciences, Changchun Normal University, Changchun, China; and Jilin Business and Technology College, Changchun, China
| | - Xueli Wang
- School of Grain, Jilin Business and Technology College, Changchun, China
| | - Yangkun Wang
- School of Life Sciences, Changchun Normal University, Changchun, China; and Jilin Business and Technology College, Changchun, China
| | - Yingqi Sun
- School of Life Sciences, Changchun Normal University, Changchun, China; and Jilin Business and Technology College, Changchun, China
| | - Yingying Su
- School of Life Sciences, Changchun Normal University, Changchun, China; and Jilin Business and Technology College, Changchun, China
| | - Xiaosong Lv
- School of Life Sciences, Changchun Normal University, Changchun, China; and Jilin Business and Technology College, Changchun, China
| | - Xiangwei Song
- School of Life Sciences, Changchun Normal University, Changchun, China; and Jilin Business and Technology College, Changchun, China
| |
Collapse
|
8
|
Zhao J, He C, Yang H, Long Y, Dong J, Wen L, Hu Z, Yin X, Hou C, Huo D. Duplex-specific nuclease powered 3D DNA walker and quantum dots barcodes for homogeneous electrochemical detection of microRNAs. Anal Chim Acta 2023; 1262:341246. [PMID: 37179061 DOI: 10.1016/j.aca.2023.341246] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/17/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023]
Abstract
Multiplex microRNAs (miRNAs) detection is beneficial for early diagnosis and prognosis of cancer. Herein, duplex-specific nuclease (DSN) powered 3D DNA walker and quantum dots (QDs) barcodes were designed for the simultaneous detection of miRNAs in a homogeneous electrochemical sensor. In the proof-of-concept experiment, the effective active area of the as-prepared graphene aerogel-modified carbon paper (CP-GAs) electrode was ∼14.30 times larger than that of the traditional glassy carbon electrode (GCE), endowing the enhanced capability of loading more metal ions for ultrasensitive detection of miRNAs. In addition, DSN-powered target recycling and DNA walking strategy assured the sensitive detection of miRNAs. After the introduction of magnetic beads (MNs) and electrochemical double enrichment strategies, the integration of triple signal amplification methods yielded good detection results. Under optimal conditions, towards simultaneous detection of microRNA-21 (miR-21) and miRNA-155 (miR-155), a linear range of 10-16-10-7 M and a sensitivity of 10 aM (miR-21) and 2.18 aM (miR-155) were achieved, respectively. It was worth mentioning that the prepared sensor can detect miR-155 down to 0.17 aM, which was also extremely advantageous among the sensors reported so far. What's more, through verification, the prepared sensor had good selectivity and reproducibility, and exhibited good detection ability in complex serum environments, showing great potential in early clinical diagnosis and screening.
Collapse
Affiliation(s)
- Jiaying Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Congjuan He
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Huisi Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Yanyi Long
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Jiangbo Dong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Li Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Zhikun Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Xinxue Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China
| | - Changjun Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China; National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Danqun Huo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, 400044, PR China; Chongqing Key Laboratory of Bio-perception & Intelligent Information Processing, School of Microelectronics and Communication Engineering, Chongqing University, Chongqing, 400044, PR China.
| |
Collapse
|
9
|
Park S, Cho E, Chueng STD, Yoon JS, Lee T, Lee JH. Aptameric Fluorescent Biosensors for Liver Cancer Diagnosis. BIOSENSORS 2023; 13:617. [PMID: 37366982 DOI: 10.3390/bios13060617] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023]
Abstract
Liver cancer is a prevalent global health concern with a poor 5-year survival rate upon diagnosis. Current diagnostic techniques using the combination of ultrasound, CT scans, MRI, and biopsy have the limitation of detecting detectable liver cancer when the tumor has already progressed to a certain size, often leading to late-stage diagnoses and grim clinical treatment outcomes. To this end, there has been tremendous interest in developing highly sensitive and selective biosensors to analyze related cancer biomarkers in the early stage diagnosis and prescribe appropriate treatment options. Among the various approaches, aptamers are an ideal recognition element as they can specifically bind to target molecules with high affinity. Furthermore, using aptamers, in conjunction with fluorescent moieties, enables the development of highly sensitive biosensors by taking full advantage of structural and functional flexibility. This review will provide a summary and detailed discussion on recent aptamer-based fluorescence biosensors for liver cancer diagnosis. Specifically, the review focuses on two promising detection strategies: (i) Förster resonance energy transfer (FRET) and (ii) metal-enhanced fluorescence for detecting and characterizing protein and miRNA cancer biomarkers.
Collapse
Affiliation(s)
- Seonga Park
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Euni Cho
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | | | - June-Sun Yoon
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Jin-Ho Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
10
|
Kang MJ, Cho YW, Kim TH. Progress in Nano-Biosensors for Non-Invasive Monitoring of Stem Cell Differentiation. BIOSENSORS 2023; 13:bios13050501. [PMID: 37232862 DOI: 10.3390/bios13050501] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/27/2023]
Abstract
Non-invasive, non-destructive, and label-free sensing techniques are required to monitor real-time stem cell differentiation. However, conventional analysis methods, such as immunocytochemistry, polymerase chain reaction, and Western blot, involve invasive processes and are complicated and time-consuming. Unlike traditional cellular sensing methods, electrochemical and optical sensing techniques allow non-invasive qualitative identification of cellular phenotypes and quantitative analysis of stem cell differentiation. In addition, various nano- and micromaterials with cell-friendly properties can greatly improve the performance of existing sensors. This review focuses on nano- and micromaterials that have been reported to improve sensing capabilities, including sensitivity and selectivity, of biosensors towards target analytes associated with specific stem cell differentiation. The information presented aims to motivate further research into nano-and micromaterials with advantageous properties for developing or improving existing nano-biosensors to achieve the practical evaluation of stem cell differentiation and efficient stem cell-based therapies.
Collapse
Affiliation(s)
- Min-Ji Kang
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Yeon-Woo Cho
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
11
|
Zheng F, Wang J, Wang D, Yang Q. Clinical Application of Small Extracellular Vesicles in Gynecologic Malignancy Treatments. Cancers (Basel) 2023; 15:cancers15071984. [PMID: 37046644 PMCID: PMC10093031 DOI: 10.3390/cancers15071984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are the key mediators of intercellular communication. They have the potential for clinical use as diagnostic or therapeutic biomarkers and have been explored as vectors for drug delivery. Identification of reliable and noninvasive biomarkers, such as sEVs, is important for early diagnosis and precise treatment of gynecologic diseases to improve patient prognosis. Previous reviews have summarized routine sEVs isolation and identification methods; however, novel and unconventional methods have not been comprehensively described. This review summarizes a convenient method of isolating sEVs from body fluids and liquid biopsy-related sEV markers for early, minimally invasive diagnosis of gynecologic diseases. In addition, the characteristics of sEVs as drug carriers and in precision treatment and drug resistance are introduced, providing a strong foundation for identifying novel and potential therapeutic targets for sEV therapy. We propose potential directions for further research on the applications of sEVs in the diagnosis and treatment of gynecologic diseases.
Collapse
|
12
|
Recent advances in plasmon-enhanced luminescence for biosensing and bioimaging. Anal Chim Acta 2023; 1254:341086. [PMID: 37005018 DOI: 10.1016/j.aca.2023.341086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/16/2023]
Abstract
Plasmon-enhanced luminescence (PEL) is a unique photophysical phenomenon in which the interaction between luminescent moieties and metal nanostructures results in a marked luminescence enhancement. PEL offers several advantages and has been extensively used to design robust biosensing platforms for luminescence-based detection and diagnostics applications, as well as for the development of many efficient bioimaging platforms, enabling high-contrast non-invasive real-time optical imaging of biological tissues, cells, and organelles with high spatial and temporal resolution. This review summarizes recent progress in the development of various PEL-based biosensors and bioimaging platforms for diverse biological and biomedical applications. Specifically, we comprehensively assessed rationally designed PEL-based biosensors that can efficiently detect biomarkers (proteins and nucleic acids) in point-of-care tests, highlighting significant improvements in the sensing performance upon the integration of PEL. In addition to discussing the merits and demerits of recently developed PEL-based biosensors on substrates or in solutions, we include a brief discussion on integrating PEL-based biosensing platforms into microfluidic devices as a promising multi-responsive detection method. The review also presents comprehensive details about the recent advances in the development of various PEL-based multi-functional (passive targeting, active targeting, and stimuli-responsive) bioimaging probes, highlighting the scope of future improvements in devising robust PEL-based nanosystems to achieve more effective diagnostic and therapeutic insights by enabling imaging-guided therapy.
Collapse
|
13
|
Tang J, Li Q, Yao C, Yang D. DNA Nanomaterial-Based Optical Probes for Exosomal miRNA Detection. Chempluschem 2023; 88:e202200345. [PMID: 36650721 DOI: 10.1002/cplu.202200345] [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: 10/06/2022] [Revised: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Micro ribonucleic acids (miRNAs) in exosomes have been proven as reliable biomarkers to detect disease progression. In recent years, deoxyribonucleic acid (DNA)-based nanomaterials show great potential in the field of diagnosis due to the programmable sequence, various molecule recognition and predictable assembly/disassembly of DNA. In this review, we focus on the molecular design and detection mechanism of DNA nanomaterials, and the developed DNA nanomaterial-based optical probes for exosomal miRNA detection are summarized and discussed. The rationally-designed DNA sequences endows these probes with low background signal and high sensitivity in exosomal miRNA detection, and the detection mechanisms based on different DNA nanomaterials are detailly introduced. At the end, the challenges and future opportunities of DNA nanomaterial-based optical probes in exosomal miRNA detection are discussed. We envision that DNA nanomaterial-based optical probes will be promising in precise biomedicine.
Collapse
Affiliation(s)
- Jianpu Tang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Qian Li
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Chi Yao
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| | - Dayong Yang
- Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), Institute of Biomolecular and Biomedical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P. R. China
| |
Collapse
|
14
|
Zhao X, He G, Deng W, Tan Y, Xie Q. Tailoring enzymatic loading capacity on 3D macroporous gold by catalytic hairpin assembly and hybridization chain reaction: Application for ultrasensitive self-powered microRNA detection. Biosens Bioelectron 2023; 219:114813. [PMID: 36270081 DOI: 10.1016/j.bios.2022.114813] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/21/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
Abstract
It is important to develop effective strategies to construct enzymatic biofuel cell based self-powered biosensors. We report here the facile regulation of enzymatic loading capacity on the bioanode by utilizing a concatenated catalytic hairpin assembly (CHA)/hybridization chain reaction (HCR) and its application for self-powered microRNA-141 (miRNA-141) detection. To construct the bioanode, a concatenated CHA/HCR process triggered by miRNA-141 was conducted on the three-dimensional macroporous gold (3DMG) electrode to generate long double-stranded DNA nanowires for glucose oxidase immobilization. Quartz crystal microbalance study reveals that the enzymatic loading capacity on the bioanode increases at an increasing miRNA-141 concentration, leading to enhanced catalytic performance for glucose oxidation. The short-circuit currents of the assembled glucose/O2 biofuel cells increase at increasing miRNA-141 concentrations, enabling ultrasensitive detection of miRNA-141. The self-powered biosensor features a wide dynamic range for detecting miRNA-141 from 10-17 to 10-11 M, with an ultralow detection limit of 1.3 aM. This work provides a highly sensitive self-powered biosensing platform for miRNA detection.
Collapse
Affiliation(s)
- Xiao Zhao
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China; State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Guihua He
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China
| | - Wenfang Deng
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China
| | - Yueming Tan
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China.
| | - Qingji Xie
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education of China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China
| |
Collapse
|
15
|
Conklin B, Conley BM, Hou Y, Chen M, Lee KB. Advanced theragnostics for the central nervous system (CNS) and neurological disorders using functional inorganic nanomaterials. Adv Drug Deliv Rev 2023; 192:114636. [PMID: 36481291 DOI: 10.1016/j.addr.2022.114636] [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: 08/30/2022] [Revised: 10/13/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Various types of inorganic nanomaterials are capable of diagnostic biomarker detection and the therapeutic delivery of a disease or inflammatory modulating agent. Those multi-functional nanomaterials have been utilized to treat neurodegenerative diseases and central nervous system (CNS) injuries in an effective and personalized manner. Even though many nanomaterials can deliver a payload and detect a biomarker of interest, only a few studies have yet to fully utilize this combined strategy to its full potential. Combining a nanomaterial's ability to facilitate targeted delivery, promote cellular proliferation and differentiation, and carry a large amount of material with various sensing approaches makes it possible to diagnose a patient selectively and sensitively while offering preventative measures or early disease-modifying strategies. By tuning the properties of an inorganic nanomaterial, the dimensionality, hydrophilicity, size, charge, shape, surface chemistry, and many other chemical and physical parameters, different types of cells in the central nervous system can be monitored, modulated, or further studies to elucidate underlying disease mechanisms. Scientists and clinicians have better understood the underlying processes of pathologies for many neurologically related diseases and injuries by implementing multi-dimensional 0D, 1D, and 2D theragnostic nanomaterials. The incorporation of nanomaterials has allowed scientists to better understand how to detect and treat these conditions at an early stage. To this end, having the multi-modal ability to both sense and treat ailments of the central nervous system can lead to favorable outcomes for patients suffering from such injuries and diseases.
Collapse
Affiliation(s)
- Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Brian M Conley
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Meizi Chen
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
16
|
Qian F, Huang Z, Zhong H, Lei Q, Ai Y, Xie Z, Zhang T, Jiang B, Zhu W, Sheng Y, Hu J, Brinker CJ. Analysis and Biomedical Applications of Functional Cargo in Extracellular Vesicles. ACS NANO 2022; 16:19980-20001. [PMID: 36475625 DOI: 10.1021/acsnano.2c11298] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Extracellular vesicles (EVs) can facilitate essential communication among cells in a range of pathophysiological conditions including cancer metastasis and progression, immune regulation, and neuronal communication. EVs are membrane-enclosed vesicles generated through endocytic origin and contain many cellular components, including proteins, lipids, nucleic acids, and metabolites. Over the past few years, the intravesicular content of EVs has proven to be a valuable biomarker for disease diagnostics, involving cancer, cardiovascular diseases, and central nervous system diseases. This review aims to provide insight into EV biogenesis, composition, function, and isolation, present a comprehensive overview of emerging techniques for EV cargo analysis, highlighting their major technical features and limitations, and summarize the potential role of EV cargos as biomarkers in disease diagnostics. Further, progress and remaining challenges will be discussed for clinical diagnostic outlooks.
Collapse
Affiliation(s)
- Feiyang Qian
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Zena Huang
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou 510970, P.R. China
| | - Hankang Zhong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Qi Lei
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yiru Ai
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Zihui Xie
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Tenghua Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Bowen Jiang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Wei Zhu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yan Sheng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - Jiaming Hu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, P.R. China
| | - C Jeffrey Brinker
- Center for Micro-Engineered Materials and the Department of Chemical and Biological Engineering, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
17
|
Park DH, Choi MY, Choi JH. Recent Development in Plasmonic Nanobiosensors for Viral DNA/RNA Biomarkers. BIOSENSORS 2022; 12:bios12121121. [PMID: 36551088 PMCID: PMC9776357 DOI: 10.3390/bios12121121] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 05/28/2023]
Abstract
Recently, due to the coronavirus pandemic, the need for early diagnosis of infectious diseases, including viruses, is emerging. Though early diagnosis is essential to prevent infection and progression to severe illness, there are few technologies that accurately measure low concentrations of biomarkers. Plasmonic nanomaterials are attracting materials that can effectively amplify various signals, including fluorescence, Raman, and other optical and electromagnetic output. In this review, we introduce recently developed plasmonic nanobiosensors for measuring viral DNA/RNA as potential biomarkers of viral diseases. In addition, we discuss the future perspective of plasmonic nanobiosensors for DNA/RNA detection. This review is expected to help the early diagnosis and pathological interpretation of viruses and other diseases.
Collapse
|
18
|
Wen Y, Zhang XW, Li YY, Chen S, Yu YL, Wang JH. Ultramultiplex NaLnF 4 Nanosatellites Combined with ICP-MS for Exosomal Multi-miRNA Analysis and Cancer Classification. Anal Chem 2022; 94:16196-16203. [DOI: 10.1021/acs.analchem.2c03727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Yun Wen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Xue-Wei Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Yuan-Yuan Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Yong-Liang Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| |
Collapse
|
19
|
Pasparakis G. Recent developments in the use of gold and silver nanoparticles in biomedicine. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1817. [PMID: 35775611 PMCID: PMC9539467 DOI: 10.1002/wnan.1817] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/18/2022]
Abstract
Gold and silver nanoparticles (NPs) are widely used in the biomedical research both in the therapeutic and the sensing/diagnostics fronts. Both metals share some common optical properties with surface plasmon resonance being the most widely exploited property in therapeutics and diagnostics. Au NPs exhibit excellent light‐to‐heat conversion efficiencies and hence have found applications primarily in precision oncology, while Ag NPs have excellent antibacterial properties which can be harnessed in biomaterials' design. Both metals constitute excellent biosensing platforms owing to their plasmonic properties and are now routinely used in various optical platforms. The utilization of Au and Ag NPs in the COVID‐19 pandemic was rapidly expanded mostly in biosensing and point‐of‐care platforms and to some extent in therapeutics. In this review article, the main physicochemical properties of Au and Ag NPs are discussed with selective examples from the recent literature. This article is categorized under:Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vitro Nanoparticle‐Based Sensing Nanotechnology Approaches to Biology > Nanoscale Systems in Biology
Collapse
Affiliation(s)
- George Pasparakis
- Department of Chemical Engineering University of Patras Patras Greece
| |
Collapse
|
20
|
Lee J, Lee JH, Mondal J, Hwang J, Kim HS, Kumar V, Raj A, Hwang SR, Lee YK. Magnetofluoro-Immunosensing Platform Based on Binary Nanoparticle-Decorated Graphene for Detection of Cancer Cell-Derived Exosomes. Int J Mol Sci 2022; 23:ijms23179619. [PMID: 36077015 PMCID: PMC9455968 DOI: 10.3390/ijms23179619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 12/13/2022] Open
Abstract
Multi-functionalized carbon nanomaterials have attracted interest owing to their excellent synergic properties, such as plasmon resonance energy transfer and surface-enhanced Raman scattering. Particularly, nanoparticle (NP)-decorated graphene (GRP) has been applied in various fields. In this study, silver NP (AgNP)- and magnetic iron oxide NP (IONP)-decorated GRP were prepared and utilized as biosensing platforms. In this case, AgNPs and GRP exhibit plasmonic properties, whereas IONPs exhibit magnetic properties; therefore, this hybrid nanomaterial could function as a magnetoplasmonic substrate for the magnetofluoro-immunosensing (MFI) system. Conversely, exosomes were recently considered high-potential biomarkers for the diagnosis of diseases. However, exosome diagnostic use requires complex isolation and purification methods. Nevertheless, we successfully detected a prostate-cancer-cell-derived exosome (PC-exosome) from non-purified exosomes in a culture media sample using Ag/IO-GRP and dye-tetraspanin antibodies (Ab). First, the anti-prostate-specific antigen was immobilized on the Ag/IO-GRP and it could isolate the PC-exosome from the sample via an external magnetic force. Dye-tetraspanin Ab was added to the sample to induce the sandwich structure. Based on the number of exosomes, the fluorescence intensity from the dye varied and the system exhibited highly sensitive and selective performance. Consequently, these hybrid materials exhibited excellent potential for biosensing platforms.
Collapse
Affiliation(s)
- Jaewook Lee
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jungpyeong 27909, Korea
- Correspondence: (J.L.); (Y.-K.L.)
| | - Ji-Heon Lee
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jungpyeong 27909, Korea
| | - Jagannath Mondal
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea
| | - Joon Hwang
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jungpyeong 27909, Korea
- Department of Aeronautical & Mechanical Design Engineering, Korea National University of Transportation, Chungju 27469, Korea
| | - Han Sang Kim
- Yonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Graduate School of Medical Science Brain Korea 21 Project, College of Medicine, Yonsei University, Seoul 03722, Korea
| | - Vinoth Kumar
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jungpyeong 27909, Korea
| | - Akhil Raj
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea
| | - Yong-Kyu Lee
- 4D Convergence Technology Institute (National Key Technology Institute in University), Korea National University of Transportation, Jungpyeong 27909, Korea
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea
- Correspondence: (J.L.); (Y.-K.L.)
| |
Collapse
|
21
|
Wu J, Wu D, Wu G, Bei HP, Li Z, Xu H, Wang Y, Wu D, Liu H, Shi S, Zhao C, Xu Y, He Y, Li J, Wang C, Zhao X, Wang S. Scale-out production of extracellular vesicles derived from natural killer cells via mechanical stimulation in a seesaw-motion bioreactor for cancer therapy. Biofabrication 2022; 14. [PMID: 35793612 DOI: 10.1088/1758-5090/ac7eeb] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/06/2022] [Indexed: 11/11/2022]
Abstract
Extracellular vesicles (EVs) derived from immune cells have shown great anti-cancer therapeutic potential. However, inefficiency in EV generation has considerably impeded the development of EV-based basic research and clinical translation. Here, we developed a seesaw-motion bioreactor (SMB) system by leveraging mechanical stimuli such as shear stress and turbulence for generating EVs with high quality and quantity from natural killer (NK) cells. Compared to EV production in traditional static culture (229 ± 74 particles per cell per day), SMB produced NK-92MI-derived EVs at a higher rate of 438 ± 50 particles per cell per day and yielded a total number of 2 × 1011 EVs over two weeks via continuous dynamic fluidic culture. In addition, the EVs generated from NK-92MI cells in SMB shared a similar morphology, size distribution, and protein profile to EVs generated from traditional static culture. Most importantly, the NK-92MI-derived EVs in SMB were functionally active in killing melanoma and liver cancer cells in both 2D and 3D culture conditions in vitro, as well as in suppressing melanoma growth in vivo. We believe that SMB is an attractive approach to producing EVs with high quality and quantity; it can additionally enhance EV production from NK92-MI cells and promote both the basic and translational research of EVs.
Collapse
Affiliation(s)
- Jianguo Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Di Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Guohua Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Ho-Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong, Hong Kong SAR, HONG KONG
| | - Zihan Li
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Han Xu
- Department of Building Environment and Energy Engineering, Xi'an Jiaotong University, 28 Xianning W Rd, Beilin, Xi'An, Shaanxi, China, 710049, Xi'an, Shanxi Province, 710049, CHINA
| | - Yimin Wang
- Institute of Translational Medicine, Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, HangZhou, 310027, CHINA
| | - Dan Wu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Hui Liu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Shengyu Shi
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, The Old Schools, Trinity Ln, Cambridge CB2 1TN, United Kingdom, Cambridge, CB2 1TN, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Yibing Xu
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Yong He
- Department of Mechanical Engineering, Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, ZheJiang, 310027, CHINA
| | - Jun Li
- Zhejiang University, 866 Yuhangtang Rd, Xihu, Hangzhou, Zhejiang, China, 310027, Hangzhou, Zhejiang Province, 310058, CHINA
| | - Changyong Wang
- Department of Neural Engineering and Biological Interdisciplinary Studies, Institude of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Academy of Military Medical Sciences, Taiping Rd. 27, 100850, Tianjin, Beijing, China, Beijing, 100850, CHINA
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Hong Kong, Hong Kong SAR, 999077, HONG KONG
| | - Shuqi Wang
- Sichuan University, 252 Shuncheng Ave, Qingyang District, Chengdu, Sichuan, China, Chengdu, Sichuan, 610017, CHINA
| |
Collapse
|
22
|
Bioprobes-regulated precision biosensing of exosomes: From the nanovesicle surface to the inside. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
23
|
Zhong Q, Huang X, Zhang R, Zhang K, Liu B. Optical Sensing Strategies for Probing Single-Cell Secretion. ACS Sens 2022; 7:1779-1790. [PMID: 35709496 DOI: 10.1021/acssensors.2c00474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Measuring cell secretion events is crucial to understand the fundamental cell biology that underlies cell-cell communication, migration, proliferation, and differentiation. Although strategies targeting cell populations have provided significant information about live cell secretion, they yield ensemble profiles that obscure intrinsic cell-to-cell variations. Innovation in single-cell analysis has made breakthroughs allowing accurate sensing of a wide variety of secretions and their release dynamics with high spatiotemporal resolution. This perspective focuses on the power of single-cell protocols to revolutionize cell-secretion analysis by allowing real-time and real-space measurements on single live cell resolution. We begin by discussing recent progress on single-cell bioanalytical techniques, specifically optical sensing strategies such as fluorescence-, surface plasmon resonance-, and surface-enhanced Raman scattering-based strategies, capable of in situ real-time monitoring of single-cell released ions, metabolites, proteins, and vesicles. Single-cell sensing platforms which allow for high-throughput high-resolution analysis with enough accuracy are highlighted. Furthermore, we discuss remaining challenges that should be addressed to get a more comprehensive understanding of secretion biology. Finally, future opportunities and potential breakthroughs in secretome analysis that will arise as a result of further development of single-cell sensing approaches are discussed.
Collapse
Affiliation(s)
- Qingmei Zhong
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Xuedong Huang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Rongrong Zhang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Kun Zhang
- Shanghai Institute for Pediatric Research, Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| |
Collapse
|
24
|
Li K, Tu J, Zhang Y, Jin D, Li T, Li J, Ni W, Xiao MM, Zhang ZY, Zhang GJ. Ultrasensitive Detection of Exosomal miRNA with PMO- Graphene Quantum Dots Functionalized Field Effect Transistor Biosensor. iScience 2022; 25:104522. [PMID: 35754714 PMCID: PMC9218366 DOI: 10.1016/j.isci.2022.104522] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/22/2022] [Accepted: 05/30/2022] [Indexed: 12/03/2022] Open
Abstract
Compared with the conventional DNA probe immobilization on the planar surface, nanoparticles-based DNA probes enable more RNA molecules to be anchored to the sensor surface, thereby improving the detection sensitivity. In this work, we report phosphorodiamidate morpholino oligomers (PMO)-graphene quantum dots (GQDs)-functionalized reduced graphene oxide (RGO) field effect transistor (FET) biosensors for ultrasensitive detection of exosomal microRNAs. After the RGO FET sensor was fabricated, polylysine (PLL) film was deposited onto the RGO surface. GQDs-PMO hybrid was prepared and covalently bound to PLL surface, enabling detection of exosomal microRNAs (miRNAs). The method achieved a detection limit as low as 85 aM and high specificity. Furthermore, the FET sensor was able to detect exosomal miRNAs in plasma samples and distinguish breast cancer samples from healthy samples. Compared with other methods, we use GQDs to further improve the sensitivity of FET, making it a potential tool for early diagnosis of breast cancer. Exosomal miRNAs are detected by GQDs-PMO-functionalized G-FET sensor The sensor can specifically detect 85 aM exosomal miRNA The results are in agreement with those of qRT-PCR This method can also distinguish breast cancer patients from healthy people
Collapse
|
25
|
Yang L, Patel KD, Rathnam C, Thangam R, Hou Y, Kang H, Lee KB. Harnessing the Therapeutic Potential of Extracellular Vesicles for Biomedical Applications Using Multifunctional Magnetic Nanomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104783. [PMID: 35132796 PMCID: PMC9344859 DOI: 10.1002/smll.202104783] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/12/2022] [Indexed: 04/14/2023]
Abstract
Extracellular vesicles (e.g., exosomes) carrying various biomolecules (e.g., proteins, lipids, and nucleic acids) have rapidly emerged as promising platforms for many biomedical applications. Despite their enormous potential, their heterogeneity in surfaces and sizes, the high complexity of cargo biomolecules, and the inefficient uptake by recipient cells remain critical barriers for their theranostic applications. To address these critical issues, multifunctional nanomaterials, such as magnetic nanomaterials, with their tunable physical, chemical, and biological properties, may play crucial roles in next-generation extracellular vesicles (EV)-based disease diagnosis, drug delivery, tissue engineering, and regenerative medicine. As such, one aims to provide cutting-edge knowledge pertaining to magnetic nanomaterials-facilitated isolation, detection, and delivery of extracellular vesicles and their associated biomolecules. By engaging the fields of extracellular vesicles and magnetic nanomaterials, it is envisioned that their properties can be effectively combined for optimal outcomes in biomedical applications.
Collapse
Affiliation(s)
- Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Piscataway, NJ 08854, USA
| | - Kapil D. Patel
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Piscataway, NJ 08854, USA
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Piscataway, NJ 08854, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Pis cataway, NJ 08854, USA
| |
Collapse
|
26
|
Jia YZ, Liu J, Wang GQ, Song ZF. miR-484: A Potential Biomarker in Health and Disease. Front Oncol 2022; 12:830420. [PMID: 35356223 PMCID: PMC8959652 DOI: 10.3389/fonc.2022.830420] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/11/2022] [Indexed: 01/30/2023] Open
Abstract
Disorders of miR-484 expression are observed in cancer, different diseases or pathological states. There is accumulating evidence that miR-484 plays an essential role in the development as well as the regression of different diseases, and miR-484 has been reported as a key regulator of common cancer and non-cancer diseases. The miR-484 targets that have effects on inflammation, apoptosis and mitochondrial function include SMAD7, Fis1, YAP1 and BCL2L13. For cancer, identified targets include VEGFB, VEGFR2, MAP2, MMP14, HNF1A, TUSC5 and KLF12. The effects of miR-484 on these targets have been documented separately. Moreover, miR-484 is typically described as an oncosuppressor, but this claim is simplistic and one-sided. This review will combine relevant basic and clinical studies to find that miR-484 promotes tumorigenesis and metastasis in liver, prostate and lung tissues. It will provide a basis for the possible mechanisms of miR-484 in early tumor diagnosis, prognosis determination, disease assessment, and as a potential therapeutic target for tumors.
Collapse
Affiliation(s)
- Yin-Zhao Jia
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Liu
- Key Laboratory of Coal Science and Technology of Ministry of Education, College of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Geng-Qiao Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Fang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Wang Y, Yang M, Shi H, Ge S, Wang X, Yu J. Photoelectrochemical Detection of Exosomal miRNAs by Combining Target-Programmed Controllable Signal Quenching Engineering. Anal Chem 2022; 94:3082-3090. [PMID: 35133793 DOI: 10.1021/acs.analchem.1c04086] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
MicroRNAs extracted from exosomes (exosomal miRNAs) have recently emerged as promising biomarkers for early prognosis and diagnosis. Thus, the development of an effective approach for exosomal miRNA monitoring has triggered extensive attention. Herein, a sensitive photoelectrochemical (PEC) biosensing platform is demonstrated for exosomal miRNA assay via the target miRNA-powered λ-exonuclease for the amplification strategy. The metal-organic framework (MOF)-decorated WO3 nanoflakes heterostructure is constructed and implemented as the photoelectrode. Also, a target exosomal miRNA-activatable programmed release nanocarrier was fabricated, which is responsible for signal control. Hemin that acted as the electron acceptor was prior entrapped into the programmed control release nanocarriers. Once the target exosomal miRNAs-21 was introduced, the as-prepared programmed release nanocarriers were initiated to trigger the release of hemin, which enabled the quenching of the photocurrent. Under the optimized conditions, the level of exosomal miRNAs-21 could be accurately tracked ranging from 1 fM to 0.1 μM with a low detection limit of 0.5 fM. The discoveries illustrate the possibility for the rapid and efficient diagnosis and prognosis prediction of diseases based on the detection of exosomal miRNAs-21 and would provide feasible approaches for the fabrication of an efficient platform for clinical applications.
Collapse
Affiliation(s)
- Yanhu Wang
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, P.R. China
| | - Mengchun Yang
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, P.R. China
| | - Huihui Shi
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| | - Shenguang Ge
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan 250022, P.R. China
| | - Xiao Wang
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, P.R. China
| | - Jinghua Yu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P.R. China
| |
Collapse
|
28
|
Chen J, Xie M, Shi M, Yuan K, Wu Y, Meng HM, Qu L, Li Z. Spatial Confinement-Derived Double-Accelerated DNA Cascade Reaction for Ultrafast and Highly Sensitive In Situ Monitoring of Exosomal miRNA and Exosome Tracing. Anal Chem 2022; 94:2227-2235. [PMID: 35029990 DOI: 10.1021/acs.analchem.1c04916] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Exosomal microRNAs (miRNAs) are reliable biomarkers of disease progression, allowing for non-invasive detection. However, detection of exosomal miRNAs in situ remains a challenge due to low abundance, poor permeability of the lipid bilayers, and slow kinetics of previous methods. Herein, an accelerated DNA nanoprobe was implemented for fast, in situ monitoring of miRNA in exosomes by employing a spatial confinement strategy. This nanoprobe not only detects miRNA in exosomes but also distinguishes tumor exosomes from those derived from normal cells with high accuracy, paving the way toward exosomal miRNA bioimaging and disease diagnosis. Furthermore, the fast response allows for this nanoprobe to be successfully utilized to monitor the process of exosomes endocytosis, making it also a tool to explore exosome biological functions.
Collapse
Affiliation(s)
- Juan Chen
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| | - Mingyue Xie
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| | - Mingqing Shi
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| | - Kun Yuan
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| | - Yanan Wu
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| | - Hong-Min Meng
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| | - Lingbo Qu
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| | - Zhaohui Li
- College of Chemistry, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Institute of Analytical Chemistry for Life Science, Zhengzhou 450001, China
| |
Collapse
|
29
|
Yoon J, Shin M, Lee JY, Lee SN, Choi JH, Choi JW. RNA interference (RNAi)-based plasmonic nanomaterials for cancer diagnosis and therapy. J Control Release 2022; 342:228-240. [PMID: 35016917 DOI: 10.1016/j.jconrel.2022.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/15/2023]
Abstract
RNA interference (RNAi) is being extensively investigated as a potential therapeutic strategy for cancer treatment. However, RNAi-based therapeutics have not yet been used to treat cancer because of their instability and the difficulty of microRNA (miRNA) delivery. Plasmonic nanoparticle-based RNAi nanotherapeutics have been developed for accurate and sensitive diagnosis and a strong therapeutic effect on cancers by leveraging their ease-of-use and specific properties such as photothermal conversion. In this review, recent strategies and advances in plasmonic nanoparticle-based miRNA delivery are briefly presented to facilitate the detection and treatment of several cancers. The challenges and potential opportunities afforded by the RNAi-based theragnosis field are discussed. We expect that the RNAi-integrated plasmonic nanotherapeutics discussed in this review can provide insights for the early diagnosis and effective treatment of cancer.
Collapse
Affiliation(s)
- Jinho Yoon
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea; Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey,123 Bevier Road, Piscataway, NJ 08854, USA
| | - Minkyu Shin
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Ji-Young Lee
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Sang-Nam Lee
- Uniance Gene Inc., 1107 Teilhard Hall, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Republic of Korea
| | - Jin-Ha Choi
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea.
| |
Collapse
|
30
|
Lu X, Yao C, Sun L, Li Z. Plasmon-enhanced biosensors for microRNA analysis and cancer diagnosis. Biosens Bioelectron 2022; 203:114041. [DOI: 10.1016/j.bios.2022.114041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 12/19/2022]
|
31
|
Min L, Wang B, Bao H, Li X, Zhao L, Meng J, Wang S. Advanced Nanotechnologies for Extracellular Vesicle-Based Liquid Biopsy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102789. [PMID: 34463056 PMCID: PMC8529441 DOI: 10.1002/advs.202102789] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 05/09/2023]
Abstract
Extracellular vesicles (EVs) are emerging as a new source of biomarkers in liquid biopsy because of their wide presence in most body fluids and their ability to load cargoes from disease-related cells. Owing to the crucial role of EVs in disease diagnosis and treatment, significant efforts have been made to isolate, detect, and analyze EVs with high efficiency. A recent overview of advanced EV detection nanotechnologies is discussed here. First, several key challenges in EV-based liquid biopsies are introduced. Then, the related pivotal advances in nanotechnologies for EV isolation based on physical features, chemical affinity, and the combination of nanostructures and chemical affinity are summarized. Next, a summary of high-sensitivity sensors for EV detection and advanced approaches for single EV detection are provided. Later, EV analysis is introduced in practical clinical scenarios, and the application of machine learning in this field is highlighted. Finally, future opportunities for the development of next-generation nanotechnologies for EV detection are presented.
Collapse
Affiliation(s)
- Li Min
- Department of GastroenterologyBeijing Friendship HospitalCapital Medical UniversityNational Clinical Research Center for Digestive DiseasesBeijing Digestive Disease CenterBeijing Key Laboratory for Precancerous Lesion of Digestive DiseaseBeijing100050P. R. China
| | - Binshuai Wang
- Department of UrologyPeking University Third HospitalBeijing100191P. R. China
| | - Han Bao
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Xinran Li
- Department of UrologyPeking University Third HospitalBeijing100191P. R. China
| | - Libo Zhao
- Echo Biotech Co., Ltd.Beijing102206P. R. China
| | - Jingxin Meng
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Shutao Wang
- Key Laboratory of Bio‐inspired Materials and Interfacial ScienceCAS Center for Excellence in NanoscienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| |
Collapse
|
32
|
Choi JH, Ha T, Shin M, Lee SN, Choi JW. Nanomaterial-Based Fluorescence Resonance Energy Transfer (FRET) and Metal-Enhanced Fluorescence (MEF) to Detect Nucleic Acid in Cancer Diagnosis. Biomedicines 2021; 9:928. [PMID: 34440132 PMCID: PMC8392676 DOI: 10.3390/biomedicines9080928] [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] [Received: 06/29/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/27/2022] Open
Abstract
Nucleic acids, including DNA and RNA, have received prodigious attention as potential biomarkers for precise and early diagnosis of cancers. However, due to their small quantity and instability in body fluids, precise and sensitive detection is highly important. Taking advantage of the ease-to-functionality and plasmonic effect of nanomaterials, fluorescence resonance energy transfer (FRET) and metal-enhanced fluorescence (MEF)-based biosensors have been developed for accurate and sensitive quantitation of cancer-related nucleic acids. This review summarizes the recent strategies and advances in recently developed nanomaterial-based FRET and MEF for biosensors for the detection of nucleic acids in cancer diagnosis. Challenges and opportunities in this field are also discussed. We anticipate that the FRET and MEF-based biosensors discussed in this review will provide valuable information for the sensitive detection of nucleic acids and early diagnosis of cancers.
Collapse
Affiliation(s)
- Jin-Ha Choi
- School of Chemical Engineering, Jeonbuk National University, Jeonju 54896, Korea;
| | - Taehyeong Ha
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (T.H.); (M.S.)
| | - Minkyu Shin
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (T.H.); (M.S.)
| | - Sang-Nam Lee
- Uniance Gene Inc., 1107 Teilhard Hall, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (T.H.); (M.S.)
| |
Collapse
|
33
|
Xiao PP, Wan QQ, Liao T, Tu JY, Zhang GJ, Sun ZY. Peptide Nucleic Acid-Functionalized Nanochannel Biosensor for the Highly Sensitive Detection of Tumor Exosomal MicroRNA. Anal Chem 2021; 93:10966-10973. [PMID: 34327982 DOI: 10.1021/acs.analchem.1c01898] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Compared with free miRNAs in blood, miRNAs in exosomes have higher abundance and stability. Therefore, miRNAs in exosomes can be regarded as an ideal tumor marker for early cancer diagnosis. Here, a peptide nucleic acid (PNA)-functionalized nanochannel biosensor for the ultrasensitive and specific detection of tumor exosomal miRNAs is proposed. After PNA was covalently bound to the inner surface of the nanochannels, the detection of tumor exosomal miRNAs was achieved by the charge changes on the surface of nanochannels before and after hybridization (PNA-miRNA). Due to the neutral characteristics of PNA, the efficiency of PNA-miRNA hybridization was improved by significantly reducing the background signal. This biosensor could not only specifically distinguish target miRNA-10b from single-base mismatched miRNA but also achieve a detection limit as low as 75 aM. Moreover, the biosensor was further used to detect exosomal miRNA-10b derived from pancreatic cancer cells and normal pancreatic cells. The results indicate that this biosensor could effectively distinguish pancreatic cancer tumor-derived exosomes from the normal control group, and the detection results show good consistency with those of the quantitative reverse-transcription polymerase chain reaction method. In addition, the biosensor was used to detect exosomal miRNA-10b in clinical plasma samples, and it was found that the content of exosomal miRNA-10b in cancer patients was generally higher than that of healthy individuals, proving that the method is expected to be applied for the early diagnosis of cancer.
Collapse
Affiliation(s)
- Ping-Ping Xiao
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, China
| | - Qiang-Qiang Wan
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, China.,Wuhan First Hospital, Wuhan 430022, China
| | - Tangbin Liao
- School of Pharmacy, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, China
| | - Ji-Yuan Tu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, China
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, China
| | - Zhong-Yue Sun
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan 430065, China
| |
Collapse
|
34
|
Zhou S, Yang Y, Wu Y, Liu S. Review: Multiplexed profiling of biomarkers in extracellular vesicles for cancer diagnosis and therapy monitoring. Anal Chim Acta 2021; 1175:338633. [PMID: 34330441 DOI: 10.1016/j.aca.2021.338633] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale vesicles secreted by normal and pathological cells. The types and levels of surface proteins and internal nucleic acids in EVs are closely related to their original cells, tumor occurrence, and development. Thus, the sensitive and accurate detection of EV biomarkers is a reliable approach for noninvasive disease diagnosis and treatment response monitoring. However, the purification and molecular profiling of these EVs are technically challenging. Much effort has been dedicated to developing new methods for the detection of multiple EV biomarkers. In this review, we summarize the recent progress in EV protein and nucleic acid biomarker analysis. Additionally, we systematically discuss the advantages of multiplexed EV biomarker detection for accurate cancer diagnosis, therapy monitoring, and cancer screening. This article aims to present an overview of all kinds of analytical technologies for assessing EVs and their applications in clinical settings.
Collapse
Affiliation(s)
- Sisi Zhou
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Yao Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China.
| | - Yafeng Wu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
35
|
Sun Z, Yang J, Li H, Wang C, Fletcher C, Li J, Zhan Y, Du L, Wang F, Jiang Y. Progress in the research of nanomaterial-based exosome bioanalysis and exosome-based nanomaterials tumor therapy. Biomaterials 2021; 274:120873. [PMID: 33989972 DOI: 10.1016/j.biomaterials.2021.120873] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 04/13/2021] [Accepted: 05/02/2021] [Indexed: 12/18/2022]
Abstract
Exosomes and their internal components have been proven to play critical roles in cell-cell interactions and intrinsic cellular regulations, showing promising prospects in both biomedical and clinical fields. Although conventional methods have so far been utilized to great effect, accurate bioanalysis remains a major challenge. In recent years, the fast-paced development of nanomaterials with unique physiochemical properties has led to a boom in the potential bioapplications of such materials. In particular, the application of nanomaterials in exosome bioanalysis provides a great opportunity to overcome the current challenges and limitations of conventional methods. A timely review of the research progress in this field is thus of great significance to the continued development of new methods. This review outlines the properties and potential uses of exosomes, and discusses the conventional methods currently used for their analysis. We then focus on exploring the current state of the art regarding the use of nanomaterials for the isolation, detection and even the subsequent profiling of exosomes. The main methods are based on principles including fluorescence, surface-enhanced Raman spectroscopy, colorimetry, electrochemistry, and surface plasmon resonance. Additionally, research on exosome-based nanomaterials tumor therapy is also promising from a clinical perspective, so the research progress in this branch is also summarized. Finally, we look at ways in which the field might develop in the future.
Collapse
Affiliation(s)
- Zhiwei Sun
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China
| | - Jingjing Yang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China
| | - Hui Li
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Cameron Fletcher
- School of Chemical Engineering, University of New South Wales, Sydney, Australia
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Yao Zhan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China; Tumor Marker Detection Engineering Technology Research Center of Shandong Province, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China.
| | - Fenglong Wang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China.
| | - Yanyan Jiang
- Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, China.
| |
Collapse
|
36
|
Wang J, Ma P, Kim DH, Liu BF, Demirci U. Towards Microfluidic-Based Exosome Isolation and Detection for Tumor Therapy. NANO TODAY 2021; 37:101066. [PMID: 33777166 PMCID: PMC7990116 DOI: 10.1016/j.nantod.2020.101066] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Exosomes are a class of cell-secreted, nano-sized extracellular vesicles with a bilayer membrane structure of 30-150 nm in diameter. Their discovery and application have brought breakthroughs in numerous areas, such as liquid biopsies, cancer biology, drug delivery, immunotherapy, tissue repair, and cardiovascular diseases. Isolation of exosomes is the first step in exosome-related research and its applications. Standard benchtop exosome separation and sensing techniques are tedious and challenging, as they require large sample volumes, multi-step operations that are complex and time-consuming, requiring cumbersome and expensive instruments. In contrast, microfluidic platforms have the potential to overcome some of these limitations, owing to their high-precision processing, ability to handle liquids at a microscale, and integrability with various functional units, such as mixers, actuators, reactors, separators, and sensors. These platforms can optimize the detection process on a single device, representing a robust and versatile technique for exosome separation and sensing to attain high purity and high recovery rates with a short processing time. Herein, we overview microfluidic strategies for exosome isolation based on their hydrodynamic properties, size filtration, acoustic fields, immunoaffinity, and dielectrophoretic properties. We focus especially on advances in label-free isolation of exosomes with active biological properties and intact morphological structures. Further, we introduce microfluidic techniques for the detection of exosomal proteins and RNAs with high sensitivity, high specificity, and low detection limits. We summarize the biomedical applications of exosome-mediated therapeutic delivery targeting cancer cells. To highlight the advantages of microfluidic platforms, conventional techniques are included for comparison. Future challenges and prospects of microfluidics towards exosome isolation applications are also discussed. Although the use of exosomes in clinical applications still faces biological, technical, regulatory, and market challenges, in the foreseeable future, recent developments in microfluidic technologies are expected to pave the way for tailoring exosome-related applications in precision medicine.
Collapse
Affiliation(s)
- Jie Wang
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Peng Ma
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Daniel H Kim
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| | - Bi-Feng Liu
- Britton Chance Center for Biomedical Photonics at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, USA
| |
Collapse
|
37
|
Yan H, Li Y, Cheng S, Zeng Y. Advances in Analytical Technologies for Extracellular Vesicles. Anal Chem 2021; 93:4739-4774. [PMID: 33635060 DOI: 10.1021/acs.analchem.1c00693] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- He Yan
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Yutao Li
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Shibo Cheng
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
| | - Yong Zeng
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States.,University of Florida Health Cancer Center, Gainesville, Florida 32610, United States
| |
Collapse
|
38
|
Choi JH, Lim J, Shin M, Paek SH, Choi JW. CRISPR-Cas12a-Based Nucleic Acid Amplification-Free DNA Biosensor via Au Nanoparticle-Assisted Metal-Enhanced Fluorescence and Colorimetric Analysis. NANO LETTERS 2021; 21:693-699. [PMID: 33346665 DOI: 10.1021/acs.nanolett.0c04303] [Citation(s) in RCA: 173] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Cell-free DNA (cfDNA) has attracted significant attention due to its high potential to diagnose diseases, such as cancer. Still, its detection by amplification method has limitations because of false-positive signals and difficulty in designing target-specific primers. CRISPR-Cas-based fluorescent biosensors have been developed but also need the amplification step for the detection. In this study, for the first time CRISPR-Cas12a based nucleic acid amplification-free fluorescent biosensor was developed to detect cfDNA by a metal-enhanced fluorescence (MEF) using DNA-functionalized Au nanoparticle (AuNP). Upon activating the CRISPR-Cas12a complex by the target cfDNA and subsequent single-strand DNA (ssDNA) degradation between AuNP and fluorophore, MEF occurred with color changes from purple to red-purple. Using this system, breast cancer gene-1 (BRCA-1) can be detected with very high sensitivity in 30 min. This rapid and highly selective sensor can be applied to measure other nucleic acid biomarkers such as viral DNA in field-deployable and point-of-care testing (POCT) platform.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Joungpyo Lim
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Minkyu Shin
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Se-Hwan Paek
- SOL Bio Corporation, Suite 510, 27, Seongsui-ro 7-gil, Seongdong-gu, Seoul 04780, Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| |
Collapse
|
39
|
Kang T, Zhu J, Luo X, Jia W, Wu P, Cai C. Controlled Self-Assembly of a Close-Packed Gold Octahedra Array for SERS Sensing Exosomal MicroRNAs. Anal Chem 2021; 93:2519-2526. [DOI: 10.1021/acs.analchem.0c04561] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tuli Kang
- Jiangsu Key Laboratory of New Power Batteries, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210097, P. R. China
| | - Jingtian Zhu
- Jiangsu Key Laboratory of New Power Batteries, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210097, P. R. China
| | - Xiaojun Luo
- Jiangsu Key Laboratory of New Power Batteries, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210097, P. R. China
| | - Wenyu Jia
- Jiangsu Key Laboratory of New Power Batteries, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210097, P. R. China
| | - Ping Wu
- Jiangsu Key Laboratory of New Power Batteries, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210097, P. R. China
| | - Chenxin Cai
- Jiangsu Key Laboratory of New Power Batteries, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210097, P. R. China
| |
Collapse
|
40
|
Suhito IR, Koo KM, Kim TH. Recent Advances in Electrochemical Sensors for the Detection of Biomolecules and Whole Cells. Biomedicines 2020; 9:15. [PMID: 33375330 PMCID: PMC7824644 DOI: 10.3390/biomedicines9010015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023] Open
Abstract
Electrochemical sensors are considered an auspicious tool to detect biomolecules (e.g., DNA, proteins, and lipids), which are valuable sources for the early diagnosis of diseases and disorders. Advances in electrochemical sensing platforms have enabled the development of a new type of biosensor, enabling label-free, non-destructive detection of viability, function, and the genetic signature of whole cells. Numerous studies have attempted to enhance both the sensitivity and selectivity of electrochemical sensors, which are the most critical parameters for assessing sensor performance. Various nanomaterials, including metal nanoparticles, carbon nanotubes, graphene and its derivatives, and metal oxide nanoparticles, have been used to improve the electrical conductivity and electrocatalytic properties of working electrodes, increasing sensor sensitivity. Further modifications have been implemented to advance sensor platform selectivity and biocompatibility using biomaterials such as antibodies, aptamers, extracellular matrix (ECM) proteins, and peptide composites. This paper summarizes recent electrochemical sensors designed to detect target biomolecules and animal cells (cancer cells and stem cells). We hope that this review will inspire researchers to increase their efforts to accelerate biosensor progress-enabling a prosperous future in regenerative medicine and the biomedical industry.
Collapse
Affiliation(s)
- Intan Rosalina Suhito
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (I.R.S.); (K.-M.K.)
| | - Kyeong-Mo Koo
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (I.R.S.); (K.-M.K.)
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (I.R.S.); (K.-M.K.)
- Integrative Research Center for Two-Dimensional Functional Materials, Institute of Interdisciplinary Convergence Research, Chung Ang University, Seoul 06974, Korea
| |
Collapse
|
41
|
Wang LL, Chen WQ, Wang YR, Zeng LP, Chen TT, Chen GY, Chen JH. Numerous long single-stranded DNAs produced by dual amplification reactions for electrochemical detection of exosomal microRNAs. Biosens Bioelectron 2020; 169:112555. [DOI: 10.1016/j.bios.2020.112555] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 02/09/2023]
|
42
|
Yoon J, Shin M, Lim J, Lee JY, Choi JW. Recent Advances in MXene Nanocomposite-Based Biosensors. BIOSENSORS 2020; 10:E185. [PMID: 33233574 PMCID: PMC7699737 DOI: 10.3390/bios10110185] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
The development of advanced biosensors with high sensitivity and selectivity is one of the most demanded concerns in the field of biosensors. To meet this requirement, up until now, numerous nanomaterials have been introduced to develop biosensors for achieving high sensitivity and selectivity. Among the latest nanomaterials attracting attention, MXene is one of the best materials for the development of biosensors because of its various superior properties. MXenes are two-dimensional inorganic compounds with few atomic layers that possess excellent characteristics including high conductivity and superior fluorescent, optical, and plasmonic properties. In this review, advanced biosensors developed on the basis of the MXene nanocomposite are discussed with the selective overview of recently reported studies. For this, introduction of the MXene including the definition, synthesis methods, and its properties are discussed. Next, MXene-based electrochemical biosensors and MXene-based fluorescent/optical biosensors are provided, which are developed on the basis of the exceptional properties of the MXene nanocomposite. This review will suggest the direction for use of the Mxene nanocomposite to develop advanced biosensors with high sensitivity and selectivity.
Collapse
Affiliation(s)
- Jinho Yoon
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea; (J.Y.); (M.S.); (J.L.)
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Minkyu Shin
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea; (J.Y.); (M.S.); (J.L.)
| | - Joungpyo Lim
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea; (J.Y.); (M.S.); (J.L.)
| | - Ji-Young Lee
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea; (J.Y.); (M.S.); (J.L.)
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea; (J.Y.); (M.S.); (J.L.)
| |
Collapse
|
43
|
Yaraki MT, Tan YN. Metal Nanoparticles-Enhanced Biosensors: Synthesis, Design and Applications in Fluorescence Enhancement and Surface-enhanced Raman Scattering. Chem Asian J 2020; 15:3180-3208. [PMID: 32808471 PMCID: PMC7693192 DOI: 10.1002/asia.202000847] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/15/2020] [Indexed: 12/17/2022]
Abstract
Metal nanoparticles (NP) that exhibit localized surface plasmon resonance play an important role in metal-enhanced fluorescence (MEF) and surface-enhanced Raman scattering (SERS). Among the optical biosensors, MEF and SERS stand out to be the most sensitive techniques to detect a wide range of analytes from ions, biomolecules to macromolecules and microorganisms. Particularly, anisotropic metal NPs with strongly enhanced electric field at their sharp corners/edges under a wide range of excitation wavelengths are highly suitable for developing the ultrasensitive plasmon-enhanced biosensors. In this review, we first highlight the reliable methods for the synthesis of anisotropic gold NPs and silver NPs in high yield, as well as their alloys and composites with good control of size and shape. It is followed by the discussion of different sensing mechanisms and recent advances in the MEF and SERS biosensor designs. This includes the review of surface functionalization, bioconjugation and (directed/self) assembly methods as well as the selection/screening of specific biorecognition elements such as aptamers or antibodies for the highly selective bio-detection. The right combinations of metal nanoparticles, biorecognition element and assay design will lead to the successful development of MEF and SERS biosensors targeting different analytes both in-vitro and in-vivo. Finally, the prospects and challenges of metal-enhanced biosensors for future nanomedicine in achieving ultrasensitive and fast medical diagnostics, high-throughput drug discovery as well as effective and reliable theranostic treatment are discussed.
Collapse
Affiliation(s)
- Mohammad Tavakkoli Yaraki
- Department of Chemical and Biomolecular EngineeringNational University of Singapore4 Engineering Drive 4Singapore117585Singapore
| | - Yen Nee Tan
- Faculty of Science, Agriculture & EngineeringNewcastle UniversityNewcastle Upon TyneNE1 7RUUnited Kingdom
- Newcastle Research & Innovation Institute (NewRIIS)80 Jurong East Street 21, #05-04 Devan Nair Institute for Employment & EmployabilitySingapore609607Singapore
| |
Collapse
|
44
|
Choi JH, Choi JW. Metal-Enhanced Fluorescence by Bifunctional Au Nanoparticles for Highly Sensitive and Simple Detection of Proteolytic Enzyme. NANO LETTERS 2020; 20:7100-7107. [PMID: 32809833 DOI: 10.1021/acs.nanolett.0c02343] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Although fluorescence-based analytical methods have been used in intracellular analyses, their sensitivity is low for the precise analysis of intracellular proteolytic enzymes to observe cell apoptosis related to cancer and neurodegenerative diseases. In this study, a metal-enhanced-fluorescence (MEF)-based highly sensitive biosensor for the detection of proteolytic enzymes is proposed for the first time by using a bifunctional Au nanoparticle (AuNP), which is connected to the fluorophore by both single-stranded DNA (ssDNA) and a peptide. Once caspase-3, a proteolytic enzyme, cuts the peptide specifically, the fluorescence signal is drastically increased because the ssDNA maintains an optimal distance for the MEF. The proposed sensing method shows the highly sensitive detection of caspase-3 based on just a simple enzymatic cleavage reaction within 1 h, and caspase-3-related preapoptotic cell detection was successfully carried out with high sensitivity. The proposed sensing method is a rapid, simple, and one-step technique for the real-time monitoring of intracellular proteolytic enzymes and can be applied to the early diagnosis of cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| |
Collapse
|
45
|
Liu P, Qian X, Li X, Fan L, Li X, Cui D, Yan Y. Enzyme-Free Electrochemical Biosensor Based on Localized DNA Cascade Displacement Reaction and Versatile DNA Nanosheets for Ultrasensitive Detection of Exosomal MicroRNA. ACS APPLIED MATERIALS & INTERFACES 2020; 12:45648-45656. [PMID: 32915531 DOI: 10.1021/acsami.0c14621] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
MicroRNA existing in exosomes (exo-miRNA) is a crucial and reliable biomarker for cancer screening and diagnosis. However, accurate detection of ultralow exo-miRNA amounts in real samples remains a challenge. Herein, a robust and ultrasensitive electrochemical biosensor was developed based on localized DNA cascade displacement reaction (L-DCDR) and versatile DNA nanosheets (DNSs) for enzyme-free analysis of exo-miRNA. The target activated L-DCDR repeatedly by consecutive toehold-mediated strand displacement, which released plentiful P strands to hybridize with capture probes immobilized on the electrode surface and DNS tags, generating an amplified electrochemical signal for the detection of exo-miRNA. The DNS could label-free load various electroactive molecules. The electrochemical biosensor revealed high sensitivity ranging from 0.1 fM to 1 nM with a limit of detection of 65 aM and good specificity. The constructed biosensor was demonstrated to be able to detect exo-miRNA derived from gastric cancer cell line (SGC-7901) and gastric cancer patients. In addition, the developed biosensor possessed several considerable advantages including simple substrate assembly, improved reaction rate, and high signal-to-noise ratio. Therefore, this strategy has great potential in bioanalysis and clinical diagnostics.
Collapse
Affiliation(s)
- Ping Liu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoqing Qian
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinmin Li
- Department of Laboratory Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400016, China
| | - Lu Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xinyu Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument National Center for Translational Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Yurong Yan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
46
|
The roles of MicroRNAs in neural regenerative medicine. Exp Neurol 2020; 332:113394. [DOI: 10.1016/j.expneurol.2020.113394] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/15/2020] [Accepted: 06/25/2020] [Indexed: 12/22/2022]
|
47
|
Cui Z, Wang XN, Lu Y, Wu P, Zhao HG, Li QL, Xu YH. miR-140 inhibits osteogenic differentiation of human periodontal ligament fibroblasts through ras homolog gene family, member A -transcriptional co-activator with PDZ-binding motif pathway. Kaohsiung J Med Sci 2020; 37:38-46. [PMID: 32841515 DOI: 10.1002/kjm2.12293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/02/2020] [Accepted: 07/19/2020] [Indexed: 12/18/2022] Open
Abstract
Osteogenesis induced by mechanical stretch is the main factor affecting the orthodontic treatment. Due to the masticatory force transmitted by tooth, human periodontal ligament fibroblasts (hPDLFs) could enhance osteogenic differentiation, and remolding of periodontal. Therefore, in-depth study of hPDLFs osteogenic differentiation and its regulatory mechanism is helpful in the understanding of periodontal remolding promoted by orthodontic force. In the present study, 3-(4,5-dimethylthiazol)-2,5-diphenyltetrazolium bromide showed that miR-140 inhibited the viability of hPDLFs cells. Moreover, we provided evidence that miR-140 inhibited alkaline phosphatase (ALP) activity, Alizarin Red S (ARS) activity and the mRNA expression of osteogenesis associated genes, including ALP, runt-related transcription factor 2, collagen 1, and osteocalcin. Besides, double-luciferase reporter result demonstrated that Ras homolog gene family, member A (RhoA) was a downstream target gene of miR-140, and by inhibiting RhoA-transcriptional co-activator with PDZ-binding motif (TAZ) signaling pathway, miR-140 suppressed the osteogenesis differentiation of hPDLFs. Furthermore, overexpression of RhoA or TAZ promoted ALP activity, ARS activity and osteogenesis associated genes expression, which was inhibited by miR-140 mimics. Our findings not only provided a possible mechanism of hPDLFs osteogenic differentiation but also proposed the clinical application of miR-140 inhibitor to target RhoA-TAZ for orthodontic treatment.
Collapse
Affiliation(s)
- Zhao Cui
- Department of General Surgery, Changchun Children's Hospital, Changchun, China
| | - Xiao-Ning Wang
- Department of Blood Transfusion, The First Hospital of Jilin University, Changchun, China
| | - Ying Lu
- Department of General Surgery, Changchun Children's Hospital, Changchun, China
| | - Peng Wu
- Department of General Surgery, Changchun Children's Hospital, Changchun, China
| | - Hong-Guang Zhao
- Department of General Surgery, Changchun Children's Hospital, Changchun, China
| | - Qiu-Lin Li
- Department of General Surgery, Changchun Children's Hospital, Changchun, China
| | - Yun-He Xu
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
48
|
Choi JH, Lee JH, Choi JW. Applications of Bionano Sensor for Extracellular Vesicles Analysis. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3677. [PMID: 32825537 PMCID: PMC7503349 DOI: 10.3390/ma13173677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
Recently, extracellular vesicles (EVs) and their contents have been revealed to play crucial roles in the intrinsic intercellular communications and have received extensive attention as next-generation biomarkers for diagnosis of diseases such as cancers. However, due to the structural nature of the EVs, the precise isolation and characterization are extremely challenging. To this end, tremendous efforts have been made to develop bionano sensors for the precise and sensitive characterization of EVs from a complex biologic fluid. In this review, we will provide a detailed discussion of recently developed bionano sensors in which EVs analysis applications were achieved, typically in optical and electrochemical methods. We believe that the topics discussed in this review will be useful to provide a concise guideline in the development of bionano sensors for EVs monitoring in the future. The development of a novel strategy to monitor various bio/chemical materials from EVs will provide promising information to understand cellular activities in a more precise manner and accelerates research on both cancer and cell-based therapy.
Collapse
Affiliation(s)
- Jin-Ha Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea;
| | - Jin-Ho Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea;
| |
Collapse
|
49
|
Lee JH, Luo J, Choi HK, Chueng STD, Lee KB, Choi JW. Functional nanoarrays for investigating stem cell fate and function. NANOSCALE 2020; 12:9306-9326. [PMID: 32090229 PMCID: PMC7671654 DOI: 10.1039/c9nr10963c] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Stem cells show excellent potential in the field of tissue engineering and regenerative medicine based on their excellent capability to not only self-renew but also differentiate into a specialized cell type of interest. However, the lack of a non-destructive monitoring system makes it challenging to identify and characterize differentiated cells before their transplantation without compromising cell viability. Thus, the development of a non-destructive monitoring method for analyzing cell function is highly desired and can significantly benefit stem cell-based therapies. Recently, nanomaterial-based scaffolds (e.g., nanoarrays) have made possible considerable advances in controlling the differentiation of stem cells and characterization of the differentiation status sensitively in real time. This review provides a selective overview of the recent progress in the synthesis methods of nanoarrays and their applications in controlling stem cell fate and monitoring live cell functions electrochemically. We believe that the topics discussed in this review can provide brief and concise guidelines for the development of novel nanoarrays and promote the interest in live cell study applications. A method which can not only control but also monitor stem cell fate and function will be a promising technology that can accelerate stem cell therapies.
Collapse
Affiliation(s)
- Jin-Ho Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Wang H, He D, Wan K, Sheng X, Cheng H, Huang J, Zhou X, He X, Wang K. In situ multiplex detection of serum exosomal microRNAs using an all-in-one biosensor for breast cancer diagnosis. Analyst 2020; 145:3289-3296. [PMID: 32255115 DOI: 10.1039/d0an00393j] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Herein, a simple all-in-one biosensor based on a DNA three-way junction has been constructed for in situ simultaneous detection of multiple miRNAs by competitive strand displacement. In our design, three oligonucleotides (Y1, Y2 and Y3) of a Y-type scaffold were extended at their 5' ends by introducing three single-stranded recognition sequences with quenchers (BHQ1, BHQ2 and BHQ2), respectively. Subsequently, three reporter sequences labeled with different fluorophores (FAM, Cy3 and Cy5) were bound to the corresponding recognition sequences to form a multicolour DNA biosensor that gives self-quenched fluorescence. The biosensor can effectively enter into exosomes and then hybridize to the complementary miRNA targets to form longer duplexes and release the reporter sequences, thus activating the readable fluorescence signals for the simultaneous detection of multiple miRNAs in exosomes. As a proof of principle, miR-21, miR-27a and miR-375 were chosen as model targets because of their high expressions in breast cancer cells (MCF-7). Fluorescence signals of MCF-7 exosomes after being treated with the biosensor exhibited positive correlations to their concentrations and the limits of detection were determined to be 0.116 μg mL-1, 0.125 μg mL-1 and 0.287 μg mL-1 for exosomes by detecting three exosomal miRNAs (miR-21, miR-27a and miR-375), respectively. In contrast, there were no obvious correlations between fluorescence intensities and control MCF-10A exosome concentrations. Importantly, by testing multiple exosomal miRNAs using the biosensor in clinical serum samples, breast cancer patients can be effectively differentiated from healthy donors. Consequently, the developed biosensor demonstrates high potential as a routine bioassay for the multiplex quantification of exosomal miRNAs in clinical diagnosis.
Collapse
Affiliation(s)
- Huizhen Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Changsha 410082, China.
| | | | | | | | | | | | | | | | | |
Collapse
|