51
|
Emerging technologies and commercial products in exosome-based cancer diagnosis and prognosis. Biosens Bioelectron 2021; 183:113176. [PMID: 33845291 DOI: 10.1016/j.bios.2021.113176] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/20/2021] [Accepted: 03/14/2021] [Indexed: 02/07/2023]
Abstract
Academic and industrial groups worldwide have reported technological advances in exosome-based cancer diagnosis and prognosis. However, the potential translation of these emerging technologies for research and clinical settings remains unknown. This work overviews the role of exosomes in cancer diagnosis and prognosis, followed by a survey on emerging exosome technologies, particularly microfluidic advances for the isolation and detection of exosomes in cancer research. The advantages and drawbacks of each of the technologies used for the isolation, detection and engineering of exosomes are evaluated to address their clinical challenges for cancer diagnosis and prognosis. Furthermore, commercial platforms for exosomal detection and analysis are introduced, and their performance and impact on cancer diagnosis and prognosis are assessed. Also, the risks associated with the further development of the next generation of exosome devices are discussed. The outcome of this work could facilitate recognizing deliverable Exo-devices and technologies with unprecedented functionality and predictable manufacturability for the next-generation of cancer diagnosis and prognosis.
Collapse
|
52
|
Peng Y, Rabin C, Walgama CT, Pollok NE, Smith L, Richards I, Crooks RM. Silver Nanocubes as Electrochemical Labels for Bioassays. ACS Sens 2021; 6:1111-1119. [PMID: 33439628 DOI: 10.1021/acssensors.0c02377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Here, we report on the use of 40 ± 4 nm silver nanocubes (AgNCs) as electrochemical labels in bioassays. The model metalloimmunoassay combines galvanic exchange (GE) and anodic stripping voltammetry (ASV). The results show that a lower limit of detection is achieved by simply changing the shape of the Ag label yielding improved GE with AgNCs when compared to GE with spherical silver nanoparticles (sAgNPs). Specifically, during GE between electrogenerated Au3+ and the Ag labels, a thin shell of Au forms on the surface of the NP. This shell is more porous when GE proceeds on AgNCs compared to sAgNPs, and therefore, more exchange occurs when using AgNCs. ASV results show that the Ag collection efficiency (AgCE%) is increased by up to ∼57% when using AgNCs. When the electrochemical system is fully optimized, the limit of detection is 0.1 pM AgNCs, which is an order of magnitude lower than that of sAgNP labels.
Collapse
Affiliation(s)
- Yi Peng
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charlie Rabin
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charuksha T. Walgama
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Nicole E. Pollok
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Leilani Smith
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Ian Richards
- Interactives Executive Excellence LLC, Austin, Texas 78733 United States
| | - Richard M. Crooks
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| |
Collapse
|
53
|
Recent advances on protein-based quantification of extracellular vesicles. Anal Biochem 2021; 622:114168. [PMID: 33741309 DOI: 10.1016/j.ab.2021.114168] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/27/2021] [Accepted: 03/06/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are secreted by almost all cells into the circulatory system and have the important function of intercellular communication. Ranging in size from 50 to 1000 nm, they are further classified based on origin, size, physical properties and function. EVs have shown the potential for studying various physiological and pathological processes, such as characterizing their parent cells with molecular markers that could further signify diseases. Proteins within EVs are the building blocks for the vesicles to function within a biological system. Isolation and proteomic profiling of EVs can advance the understanding of their biogenesis and functions, which can give further insight of how they can be used in clinical settings. However, the nanoscale size of EVs, which is much smaller than that of cells, comprises a major challenge for EV isolation and the characterization of their protein cargos. With the recent advances of bioanalytical techniques such as lab-on-a-chip devices and innovated flow cytometry, the quantification of EV proteins from a small number of vesicles down to the single vesicle level has been achieved, shining light on the promising applications of these small vesicles for early disease diagnosis and treatment monitoring. In this article, we first briefly review conventional EV protein determination technologies and their limitations, followed by detailed description and analysis of emerging technologies used for EV protein quantification, including optical, non-optical, microfluidic, and single vesicle detection methods. The pros and cons of these technologies are compared and the current challenges are outlined. Future perspectives and potential research directions of the EV protein analysis methods are discussed.
Collapse
|
54
|
Zubair M, Wang S, Ali N. Advanced Approaches to Breast Cancer Classification and Diagnosis. Front Pharmacol 2021; 11:632079. [PMID: 33716731 PMCID: PMC7952319 DOI: 10.3389/fphar.2020.632079] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022] Open
Abstract
The International Agency for Research on Cancer (IARC) has recently reported a 66% increase in the global number of cancer deaths since 1960. In the US alone, about one in eight women is expected to develop invasive breast cancer(s) (breast cancer) at some point in their lifetime. Traditionally, a BC diagnosis includes mammography, ultrasound, and some high-end molecular bioimaging. Unfortunately, these techniques detect BC at a later stage. So early and advanced molecular diagnostic tools are still in demand. In the past decade, various histological and immuno-molecular studies have demonstrated that BC is highly heterogeneous in nature. Its growth pattern, cytological features, and expression of key biomarkers in BC cells including hormonal receptor markers can be utilized to develop advanced diagnostic and therapeutic tools. A cancer cell's progression to malignancy exhibits various vital biomarkers, many of which are still underrepresented in BC diagnosis and treatment. Advances in genetics have also enabled the development of multigene assays to detect genetic heterogeneity in BC. However, thus far, the FDA has approved only four such biomarkers-cancer antigens (CA); CA 15-3, CA 27-29, Human epidermal growth factor receptor 2 (HER2), and circulating tumor cells (CTC) in assessing BC in body fluids. An adequately structured portable-biosensor with its non-invasive and inexpensive point-of-care analysis can quickly detect such biomarkers without significantly compromising its specificity and selectivity. Such advanced techniques are likely to discriminate between BC and a healthy patient by accurately measuring the cell shape, structure, depth, intracellular and extracellular environment, and lipid membrane compositions. Presently, BC treatments include surgery and systemic chemo- and targeted radiation therapy. A biopsied sample is then subjected to various multigene assays to predict the heterogeneity and recurrence score, thus guiding a specific treatment by providing complete information on the BC subtype involved. Thus far, we have seven prognostic multigene signature tests for BC providing a risk profile that can avoid unnecessary treatments in low-risk patients. Many comparative studies on multigene analysis projected the importance of integrating clinicopathological information with genomic-imprint analysis. Current cohort studies such as MINDACT, TAILORx, Trans-aTTOM, and many more, are likely to provide positive impact on long-term patient outcome. This review offers consolidated information on currently available BC diagnosis and treatment options. It further describes advanced biomarkers for the development of state-of-the-art early screening and diagnostic technologies.
Collapse
Affiliation(s)
- M. Zubair
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - S. Wang
- Department of Chemistry, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - N. Ali
- Department of Biology, University of Arkansas at Little Rock, Little Rock, AR, United States
| |
Collapse
|
55
|
Zhang H, Qiao B, Guo Q, Jiang J, Cai C, Shen J. A facile and label-free electrochemical aptasensor for tumour-derived extracellular vesicle detection based on the target-induced proximity hybridization of split aptamers. Analyst 2021; 145:3557-3563. [PMID: 32309839 DOI: 10.1039/d0an00066c] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Facile detection of tumour-derived extracellular vesicles (EVs) is crucial to cancer diagnosis. Herein, a facile and label-free electrochemical aptasensor was fabricated to detect tumour-derived EVs based on the target-induced proximity hybridization of split aptamers. In this assay, two designed oligonucleotide probes containing fragments of a protein tyrosine kinase-7 (PTK7) aptamer were used to recognize and capture EVs containing PTK7. In the presence of target EVs, the aptamer-target ternary complex could induce proximity hybridization and form a DNA duplex on the electrode. The DNA duplex could bind more electroactive Ru(NH3)63+ through electrostatic attraction, resulting in an increased cathodic current signal. By virtue of the excellent electrochemical signal reporter RuHex, the specificity of the aptamer and proximity ligation, a facile EV electrochemical aptasensor with a detection limit of 6.607 × 105 particles per mL was realized. Furthermore, this aptasensor showed good selectivity to distinguish different tumour-derived EVs and was applied to detect EVs in complex biological samples. The proposed electrochemical aptasensor can be further extended to the detection of other EVs, thus showing great potential in clinical diagnosis.
Collapse
Affiliation(s)
- Hui Zhang
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Bin Qiao
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Qunqun Guo
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Juqian Jiang
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Chenxin Cai
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Jian Shen
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| |
Collapse
|
56
|
Dell'Olio F, Su J, Huser T, Sottile V, Cortés-Hernández LE, Alix-Panabières C. Photonic technologies for liquid biopsies: recent advances and open research challenges. LASER & PHOTONICS REVIEWS 2021; 15:2000255. [PMID: 35360260 PMCID: PMC8966629 DOI: 10.1002/lpor.202000255] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Indexed: 05/15/2023]
Abstract
The recent development of sophisticated techniques capable of detecting extremely low concentrations of circulating tumor biomarkers in accessible body fluids, such as blood or urine, could contribute to a paradigm shift in cancer diagnosis and treatment. By applying such techniques, clinicians can carry out liquid biopsies, providing information on tumor presence, evolution, and response to therapy. The implementation of biosensing platforms for liquid biopsies is particularly complex because this application domain demands high selectivity/specificity and challenging limit-of-detection (LoD) values. The interest in photonics as an enabling technology for liquid biopsies is growing owing to the well-known advantages of photonic biosensors over competing technologies in terms of compactness, immunity to external disturbance, and ultra-high spatial resolution. Some encouraging experimental results in the field of photonic devices and systems for liquid biopsy have already been achieved by using fluorescent labels and label-free techniques and by exploiting super-resolution microscopy, surface plasmon resonance, surface-enhanced Raman scattering, and whispering gallery mode resonators. This paper critically reviews the current state-of-the-art, starting from the requirements imposed by the detection of the most common circulating biomarkers. Open research challenges are considered together with competing technologies, and the most promising paths of improvement are discussed for future applications.
Collapse
Affiliation(s)
- Francesco Dell'Olio
- Department of Electrical and Information Engineering, Polytechnic University of Bari, 70125, Italy
| | - Judith Su
- Department of Biomedical Engineering, College of Optical Sciences, and BIO5 Institute, University of Arizona, 85721, USA
| | - Thomas Huser
- Biomolecular Photonics, Department of Physics, University of Bielefeld, 33615 Germany
| | - Virginie Sottile
- Department of Molecular Medicine, University of Pavia, 27100, Italy
| | | | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Center of Montpellier, 34093 CEDEX 5, France
| |
Collapse
|
57
|
Shi X, Chen L, Chen S, Sun D. Electrochemical aptasensors for the detection of hepatocellular carcinoma-related biomarkers. NEW J CHEM 2021. [DOI: 10.1039/d1nj01042e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent progress in electrochemical aptasensors for the detection of HCC-related biomarkers, including cancer cells, proteins, cell-derived exosomes, and nucleic acids, is reviewed.
Collapse
Affiliation(s)
- Xianhua Shi
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Linxi Chen
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Siyi Chen
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Duanping Sun
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| |
Collapse
|
58
|
Tiwari S, Kumar V, Randhawa S, Verma SK. Preparation and characterization of extracellular vesicles. Am J Reprod Immunol 2020; 85:e13367. [PMID: 33118232 DOI: 10.1111/aji.13367] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous membranous vesicles secreted by every cell type and offer significant potential in therapy and diagnostics. Differential ultracentrifugation is the gold standard for EV isolation, although other techniques including, polyethylene glycol (PEG) precipitation, immunoprecipitation, size exclusion chromatography, and immuno-isolation approaches are common. Purified EVs can be characterized based on their physical characteristics, biochemical composition, or cell of origin. For size and concentration measurement, nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), and electron microscopy are commonly employed methods. Biochemical analyses of EVs are typically performed using flow cytometry, immunoblotting, or proteomic investigation. Based on tissue of origin, EVs have specific markers that can be used to isolate and purify specific cell-associated EVs using an affinity selection approach. Despite existence of several methods for isolation and characterization, major limitations associated with each method hinder the progress of the field. Evolving concepts in EV biology possess great promise for better isolation and characterization leading to a better insight of biological function and have immense clinical implications. In this review, we discuss recent advancements in EV isolation and characterization approaches.
Collapse
Affiliation(s)
- Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi PGI, Lucknow, India
| | - Vinod Kumar
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi PGI, Lucknow, India
| | | | - Santosh K Verma
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi PGI, Lucknow, India
| |
Collapse
|
59
|
Recent advances in nanomaterial-based biosensors for the detection of exosomes. Anal Bioanal Chem 2020; 413:83-102. [PMID: 33164151 DOI: 10.1007/s00216-020-03000-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 12/24/2022]
Abstract
Exosomes are a type of extracellular vesicle actively secreted by almost all eukaryotic cells. They are ideal candidates for reliable next-generation biomarkers in the early diagnosis and therapeutic response evaluation of cancer. Thus, the quantification of exosomes is crucial in facilitating clinical research and application. Compared with traditional materials, nanomaterials have better optical, magnetic, electrical, and catalytic properties due to their small size, high specific surface area, and variable structure. The incorporation of nanomaterials into sensing systems is an attractive approach towards improving sensitivity and can provide improved sensor selectivity and stability. In this paper, we summarize the progress in nanomaterial-based exosome detection methods, including electrochemical biosensors, photoelectrochemical biosensors, colorimetric biosensors, fluorescence biosensors, chemiluminescence biosensors, electrochemiluminescence biosensors, surface plasmon resonance biosensors, and surface-enhanced Raman spectroscopy biosensors. Moreover, future research directions and challenges in exosome detection methods are discussed. We hope that this article will offer an overview of nanomaterial-based exosome detection techniques and open new avenues in disease research.Graphical abstract.
Collapse
|
60
|
Shen M, Di K, He H, Xia Y, Xie H, Huang R, Liu C, Yang M, Zheng S, He N, Li Z. Progress in exosome associated tumor markers and their detection methods. MOLECULAR BIOMEDICINE 2020; 1:3. [PMID: 35006428 PMCID: PMC8603992 DOI: 10.1186/s43556-020-00002-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023] Open
Abstract
Exosomes are secreted by cells and are widely present in body fluids. Exosomes contain various molecular constituents of their cells of origin such as proteins, mRNA, miRNAs, DNA, lipid and glycans which are very similar as the content in tumor cells. These contents play an important role in various stages of tumor development, and make the tumor-derived exosome as a hot and emerging biomarker for various cancers diagnosis and management in non-invasive manner. The present problems of exosome isolation and detection hinder the application of exosomes. With the development of exosome isolation and detection technology, the contents of exosomes can be exploited for early cancer diagnosis. This review summarizes the recent progress on exosome-associated tumor biomarkers and some new technologies for exosome isolation and detection. Furthermore, we have also discussed the future development direction in exosome analysis methods.
Collapse
Affiliation(s)
- Mengjiao Shen
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Shanghai Health Development Research Center, Shanghai, China
| | - Kaili Di
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hongzhang He
- Captis Diagnostics Inc, Pittsburgh, PA, 15213, USA
| | - Yanyan Xia
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hui Xie
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Rongrong Huang
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Chang Liu
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mo Yang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hunghom, Kowloon, Hong Kong, People's Republic of China.
| | - Siyang Zheng
- Department of Biomedical Engineering and Electrical & Computer Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Scott Hall 4N211, Pittsburgh, PA, 15213, USA.
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Zhiyang Li
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
61
|
Masaoutis C, Al Besher S, Koutroulis I, Theocharis S. Exosomes in Nephropathies: A Rich Source of Novel Biomarkers. DISEASE MARKERS 2020; 2020:8897833. [PMID: 32849923 PMCID: PMC7441435 DOI: 10.1155/2020/8897833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/08/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
The biomarkers commonly utilized in diagnostic evaluations of kidney disease suffer from low sensitivity, especially in the early stages of renal damage. On the other hand, obtaining a renal biopsy to augment clinical decision making can lead to potentially serious complications. In order to overcome the shortcomings of currently available diagnostic tools, recent studies suggest that exosomes, cell-secreted extracellular vesicles containing a large array of active molecules to facilitate cell-to-cell communication, may represent a rich source of novel disease biomarkers. Because of their endocytic origin, exosomes carry markers typical for their parent cells, which could permit the localization of biochemical cellular alterations in specific kidney compartments. Different types of exosomes can be isolated from noninvasively obtained biofluids; however, in the context of kidney disease, evidence has emerged on the role of urinary exosomes in the diagnostic and predictive modeling of renal pathology. The current review summarizes the potential application of exosomes in the detection of acute and chronic inflammatory, metabolic, degenerative, and genetic renal diseases.
Collapse
Affiliation(s)
- Christos Masaoutis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias street, Bld 10, Goudi, 11527 Athens, Greece
| | - Samer Al Besher
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias street, Bld 10, Goudi, 11527 Athens, Greece
| | - Ioannis Koutroulis
- Children's National Hospital, Division of Emergency Medicine and Center for Genetic Medicine, George Washington University School of Medicine and Health Sciences, 111 Michigan Ave. NW, Washington, DC 20010, USA
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias street, Bld 10, Goudi, 11527 Athens, Greece
| |
Collapse
|
62
|
Yu X, Sha L, Dong L, Cao Y, Zhao J. Recent Advances in Bio-Sensing Methods for the Detection of Tumor Exosomes. Crit Rev Anal Chem 2020; 52:356-374. [PMID: 32762253 DOI: 10.1080/10408347.2020.1802220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Exosomes, small vesicles with the diameters of 40-160 nm, play an important role in intercellular transport and communication. Exosomes are rich in many kinds of biomolecules, and differential expression of exosomal contents directly reflects the state of the original cells. Therefore, the tumor exosomes are appearing as promising biomarkers in liquid biopsy, and highly sensitive and specific detection of tumor exosomes may provide the information for the early diagnosis, real-time monitoring and treatment of the tumors. In this review, we summarized the recent advances in the detection of tumor exosomes, mainly focusing on the use of different analytical techniques, such as optical and electrochemical methods as well as that combination with newly-emerging microfluidic techniques, thereby providing valuable information for the application in the clinical diagnosis and management of the tumors.
Collapse
Affiliation(s)
- Xiaomeng Yu
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Lingjun Sha
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Langjian Dong
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Ya Cao
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, P. R. China
| | - Jing Zhao
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, P. R. China
| |
Collapse
|
63
|
Yu Y, Zhang WS, Guo Y, Peng H, Zhu M, Miao D, Su G. Engineering of exosome-triggered enzyme-powered DNA motors for highly sensitive fluorescence detection of tumor-derived exosomes. Biosens Bioelectron 2020; 167:112482. [PMID: 32795917 DOI: 10.1016/j.bios.2020.112482] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023]
Abstract
Tumor-derived exosomes containing multiple proteins originating from parent cancer cells have emerged as biomarkers for cancer diagnosis. Herein, we propose a three-dimensional DNA motor-based exosome assay platform for the selective and sensitive detection of exosomes. The DNA motor used gold nanoparticle (GNP) tracks, consisting of fluorescein-labeled substrate strands and aptamer-locked motor strands. Recognition of the target protein on exosomes by its aptamer unlocked the motor strand and triggered the DNA motor process. Powered by restriction endonuclease, the motor strands autonomously walked along the GNP track. Each movement step cleaved one substrate strand and restored one fluorescein molecule. For exosome detection, the proposed method displayed a broad dynamic range acrossing 5 orders of magnitude with the detection limit as low as 8.2 particles/μL in PBS. The method also exhibited good selectivity among different tumor-derived exosomes and performed well in complex biological samples. The capability to profile exosomal surface proteins efficiently endowed our DNA motor great potential for developing a simple and cost-effective device for clinical diagnosis.
Collapse
Affiliation(s)
- Yanyan Yu
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Wei S Zhang
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Yuehua Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Huaping Peng
- School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
| | - Min Zhu
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Dandan Miao
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, 226001, China.
| |
Collapse
|
64
|
Zhang M, Vojtech L, Ye Z, Hladik F, Nance E. Quantum Dot Labeling and Visualization of Extracellular Vesicles. ACS APPLIED NANO MATERIALS 2020; 3:7211-7222. [PMID: 34568770 PMCID: PMC8460064 DOI: 10.1021/acsanm.0c01553] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Extracellular vesicles (EVs) are important mediators of intercellular communication. Their role in disease processes, uncovered mostly over the last two decades, makes them potential biomarkers, leading to a need to fundamentally understand EV biology. Direct visualization of EVs can provide insights into EV behavior, but current labeling techniques are often restricted by false-positive signals and rapid photobleaching. Hence, we developed a method of labeling EVs through conjugation with quantum dots (QDs)-high photoluminescent nanosized semi-conductors-using click chemistry. We showed that QD-EV conjugation could be tailored by altering QD to EV ratio or by using a catalyst. This conjugation chemistry was stable in a biological environment and upon storage for up to a week. Using size-exclusion chromatography, QD-EV conjugates could be separated from unconjugated QDs, enabling EV-specific signal detection. We demonstrate that these QD-EV conjugates can be live- and fixed-imaged in high resolution on cells and in tissue sheets, and the conjugates have better photostability compared with the commonly used EV dye DiI. We labeled two distinct EV populations: human semen EVs (sEVs) from fresh semen samples donated by healthy volunteers and brain EVs (bEVs) from excised rat brain tissues. We visualized QD-sEVs in epithelial sheets isolated from human vaginal mucosa and time-lapse imaged QD-bEV interactions with microglial BV-2 cells. The development of the QD-EV conjugate will benefit the study of EV localization, movement, and function and accelerate their potential use as biomarkers, therapeutic agents, or drug-delivery vehicles.
Collapse
Affiliation(s)
- Mengying Zhang
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington 98195-1652, United States
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington 98195-6460, United States
| | - Ziming Ye
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195-1750, United States
| | - Florian Hladik
- Department of Obstetrics and Gynecology and Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington 98195-6460, United States
| | - Elizabeth Nance
- Molecular Engineering and Sciences Institute, Department of Chemical Engineering, Center on Human Development and Disability, and Department of Radiology, University of Washington, Seattle, Washington 98195-1652, United States
| |
Collapse
|
65
|
Tang Z, Huang J, He H, Ma C, Wang K. Contributing to liquid biopsy: Optical and electrochemical methods in cancer biomarker analysis. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213317] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
66
|
Ohannesian N, Gunawardhana L, Misbah I, Rakhshandehroo M, Lin SH, Shih WC. Commercial and emerging technologies for cancer diagnosis and prognosis based on circulating tumor exosomes. JPHYS PHOTONICS 2020. [DOI: 10.1088/2515-7647/ab8699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Abstract
Exosomes are nano-sized extracellular vesicles excreted by mammalian cells that circulate freely in the bloodstream of living organisms. Exosomes have a lipid bilayer that encloses genetic material used in intracellular communication (e.g. double-stranded DNA, micro-RNAs, and messenger RNA). Recent evidence suggests that dysregulation of this genetic content within exosomes has a major role in tumor progression in the surrounding microenvironment. Motivated by this discovery, we focused here on using exosomal biomarkers as a diagnostic and prognostic tool for cancer. In this review, we discuss recently discovered exosome-derived proteomic and genetic biomarkers used in cancer diagnosis and prognosis. Although several genetic biomarkers have been validated for their diagnostic values, proteomic biomarkers are still being actively pursued. We discuss both commercial technologies and emerging technologies for exosome isolation and analysis. Emerging technologies can be classified into optical and non-optical methods. The working principle of each method is briefly discussed as well as advantages and limitations.
Collapse
|
67
|
Kholafazad Kordasht H, Hasanzadeh M. Biomedical analysis of exosomes using biosensing methods: recent progress. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2020; 12:2795-2811. [PMID: 32930202 DOI: 10.1039/d0ay00722f] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Exosomes are membrane-bound extracellular vesicles (EVs) that are produced in the endosomal compartments of most eukaryotic cells; they play important roles in intercellular communication in diverse cellular processes and transmit different types of biomolecules. Endocytic pathways release exosomes, which have diameters ranging from 50 to 200 nm. The unique functions of exosomes have been introduced as cancer bio-markers due to the cargo (protein, DNA and RNA) of external exosomes (tetraspanin) and internal exosomes (syntenin). The early detection of cancer by exosomes can be an excellent method for the treatment of cancer. Although detection methods based on exosomes are important, they require extensive sample purification, have high false-positive rates, and encounter labeling difficulties due to the small size of exosomes. Here, we have reviewed three major types of biosensors, namely, electrochemical biosensors, optical biosensors and electrochemiluminescence biosensors for the detection of exosomes released from breast, ovarian, pancreatic, lung, and cervical cancer cells. In addition, the importance of nanomaterials and their applications in the biomedical analysis of exosomes are discussed. Although exosomes can be used to identify various types of external and internal biomarkers by conjugating with recognition elements, most designed biosensors are based on CD9 and CD63. Therefore, the development of novel biosensors for the selective and sensitive detection of exosomes is a current challenge. We hope that this review will serve as a beneficial study for improving exosome detection in clinical samples.
Collapse
Affiliation(s)
- Houman Kholafazad Kordasht
- Department of Food Hygiene and Aquatic, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hasanzadeh
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Food and Drug Safety Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
68
|
Selected Tetraspanins Functionalized Niosomes as Potential Standards for Exosome Immunoassays. NANOMATERIALS 2020; 10:nano10050971. [PMID: 32443605 PMCID: PMC7712311 DOI: 10.3390/nano10050971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/26/2020] [Accepted: 05/08/2020] [Indexed: 02/07/2023]
Abstract
Quantitative detection of exosomes in bio-fluids is a challenging task in a dynamic research field. The absence of a well-established reference material (RM) for method development and inter-comparison studies could be potentially overcome with artificial exosomes: lab-produced biomimetic particles with morphological and functional properties close to natural exosomes. This work presents the design, development and functional characteristics of fully artificial exosomes based on tetraspanin extracellular loops-coated niosomes, produced by bio-nanotechnology methods based on supra-molecular chemistry and recombinant protein technology. Mono- and double-functionalized particles with CD9/CD63 tetraspanins have been developed and characterized from a morphological and functional point of view. Produced bio-particles showed close similarities with natural entities in terms of physical properties. Their utility for bioanalysis is demonstrated by their detection and molecular-type discrimination by enzyme-linked immunosorbent assays (ELISAs), one of the most frequent bio-analytical method found in routine and research labs. The basic material based on streptavidin-coated niosomes allows the surface functionalization with any biotinylated protein or peptide, introducing versatility. Although promising results have been reported, further optimizations and deeper characterization will help this innovative biomaterial become a robust RM for validation and development of diagnostic tools for exosomes determination.
Collapse
|
69
|
Huang Z, Lin Q, Ye X, Yang B, Zhang R, Chen H, Weng W, Kong J. Terminal deoxynucleotidyl transferase based signal amplification for enzyme-linked aptamer-sorbent assay of colorectal cancer exosomes. Talanta 2020; 218:121089. [PMID: 32797865 DOI: 10.1016/j.talanta.2020.121089] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022]
Abstract
Exosomes have received increasingly significant attention and have shown great clinical value as biomarkers for a number of diseases. However, there is still a lack of a highly sensitive and visualized method for the detection of exosomes in numerous samples simultaneously. Here, we developed a high-throughput, colorimetric and simple method to detect colorectal cancer (CRC) exosomes based on terminal deoxynucleotidyl transferase (TdT)-aided ultraviolet signal amplification. Anti-A33, a CRC exosomal protein marker, was selected as a capture probe, and a facility-prepared EpCAM (CRC exosomal protein) aptamer-Au-primer complex was used as a signal probe. After the CRC exosomes were captured onto the surface of 96-well plates, the primer was extended to the poly(biotin-adenine) chains with the help of TdT, resulting in an increase in the binding amount of avidin-modified horseradish peroxidase (Av-HRP) for H2O2-mediated oxidation of 3,3',5,5'-tetramethyl benzidine (TMB) in enzyme-linked aptamer-sorbent assay (ELASA). The results showed that the incorporation of ploy(biotin-A) enabled approximately 10.4-fold signal amplification. This approach achieved a linear range of 9.75 × 103-1.95 × 106 particles/μL for CRC cell-derived exosomes. The feasibility of the developed assay was evaluated using clinical serum samples. CRC patients (n = 16) could be clearly and successfully distinguished from healthy individuals (n = 9). Furthermore, this proposed platform holds considerable potential for the detection of different targets, simply by changing the aptamer and antibody.
Collapse
Affiliation(s)
- Zhipeng Huang
- Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Qiuyuan Lin
- Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Xin Ye
- Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Bin Yang
- Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Ren Zhang
- Department of Chemistry, Fudan University, Shanghai, 200438, China
| | - Hui Chen
- Department of Chemistry, Fudan University, Shanghai, 200438, China.
| | - Wenhao Weng
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China.
| | - Jilie Kong
- Department of Chemistry, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
70
|
Tran PHL, Xiang D, Tran TTD, Yin W, Zhang Y, Kong L, Chen K, Sun M, Li Y, Hou Y, Zhu Y, Duan W. Exosomes and Nanoengineering: A Match Made for Precision Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904040. [PMID: 31531916 DOI: 10.1002/adma.201904040] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/02/2019] [Indexed: 05/28/2023]
Abstract
Targeted exosomal delivery systems for precision nanomedicine attract wide interest across areas of molecular cell biology, pharmaceutical sciences, and nanoengineering. Exosomes are naturally derived 50-150 nm nanovesicles that play important roles in cell-to-cell and/or cell-to-tissue communications and cross-species communication. Exosomes are also a promising class of novel drug delivery vehicles owing to their ability to shield their payload from chemical and enzymatic degradations as well as to evade recognition by and subsequent removal by the immune system. Combined with a new class of affinity ligands known as aptamers or chemical antibodies, molecularly targeted exosomes are poised to become the next generation of smartly engineered nanovesicles for precision medicine. Here, recent advances in targeted exosomal delivery systems engineered by aptamer for future strategies to promote human health using this class of human-derived nanovesicles are summarized.
Collapse
Affiliation(s)
- Phuong H L Tran
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
| | - Dongxi Xiang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, 77 Avenue Louise Pasteur, Boston, MA, 02115, USA
| | - Thao T D Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Wang Yin
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
| | - Yumei Zhang
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
| | - Lingxue Kong
- Institute for Frontier Materials, Deakin University, Geelong, Victoria, 3216, Australia
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, He'nan Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, He'nan Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, and St George and Sutherland Clinical School, University of New South Wales, Kensington, NSW, 2217, Australia
| | - Yingchun Hou
- Laboratory of Tumor Molecular and Cellular Biology, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi, 710119, China
| | - Yimin Zhu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
- GenePharma-Deakin Joint Laboratory of Aptamer Medicine, Suzhou, 215123, China
- GenePharma-Deakin Joint Laboratory of Aptamer Medicine, Waurn Ponds, Victoria, 3216, Australia
| |
Collapse
|
71
|
Xu L, Shoaie N, Jahanpeyma F, Zhao J, Azimzadeh M, Al Jamal KT. Optical, electrochemical and electrical (nano)biosensors for detection of exosomes: A comprehensive overview. Biosens Bioelectron 2020; 161:112222. [PMID: 32365010 DOI: 10.1016/j.bios.2020.112222] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
Exosomes are small extracellular vesicles involved in many physiological activities of cells in the human body. Exosomes from cancer cells have great potential to be applied in clinical diagnosis, early cancer detection and target identification for molecular therapy. While this field is gaining increasing interests from both academia and industry, barriers such as supersensitive detection techniques and highly-efficient isolation methods remain. In the clinical settings, there is an urgent need for rapid analysis, reliable detection and point-of-care testing (POCT). With these challenges to be addressed, this article aims to review recent developments and technical breakthroughs including optical, electrochemical and electrical biosensors for exosomes detection in the field of cancer and other diseases and demonstrate how nanobiosensors could enhance the performance of conventional sensors. Working strategies, limit of detections, advantages and shortcomings of the studies are summarized. New trends, challenges and future perspectives of exosome-driven POCT in liquid biopsy have been discussed.
Collapse
Affiliation(s)
- Lizhou Xu
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Nahid Shoaie
- Department of Biotechnology, Tarbiat Modares University of Medical Science, Tehran, Iran
| | - Fatemeh Jahanpeyma
- Department of Biotechnology, Tarbiat Modares University of Medical Science, Tehran, Iran
| | - Junjie Zhao
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom
| | - Mostafa Azimzadeh
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, 89195-999, Yazd, Iran; Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, 89195-999, Yazd, Iran; Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, 8916188635, Yazd, Iran.
| | - Khuloud T Al Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, United Kingdom.
| |
Collapse
|
72
|
Liao G, Liu X, Yang X, Wang Q, Geng X, Zou L, Liu Y, Li S, Zheng Y, Wang K. Surface plasmon resonance assay for exosomes based on aptamer recognition and polydopamine-functionalized gold nanoparticles for signal amplification. Mikrochim Acta 2020; 187:251. [PMID: 32232575 DOI: 10.1007/s00604-020-4183-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
A novel surface plasmon resonance (SPR) strategy is introduced for the specific determination of exosomes based on aptamer recognition and polydopamine-functionalized gold nanoparticle (Au@PDA NP)-assisted signal amplification. Exosomes derived from hepatic carcinoma SMMC-7721 were selected as the model target. SMMC-7721 exosomes can be specifically captured by the aptamer ZY-sls that was complementary to the DNA tetrahedron probes (DTPs), and then the CD63 aptamer-linked Au@PDA NPs recognized SMMC-7721 exosomes for signal amplification. The DTPs were modified on a Au film for preventing Au deposition on the surface during the introduction of HAuCl4, and PDA coated on the AuNPs was used to reduce HAuCl4 in situ without any reductant assistance. It results in a further enhanced SPR signal. The assay can clearly distinguish SMMC-7721 exosomes from others (HepG2 exosomes, Bel-7404 exosomes, L02 exosomes, MCF-7 exosomes, and SW480 exosomes, respectively). SMMC-7721 exosomes are specifically determined as low as 5.6 × 105 particles mL-1. The method has successfully achieved specific determination of SMMC-7721 exosomes even in 50% of human serum without any pretreatment. Graphical abstract A novel surface plasmon resonance (SPR) strategy was introduced for the specific determination of exosomes based on aptamer recognition and polydopamine functionalized gold nanoparticles (Au@PDA NPs). The SPR signal was improved using the Au@PDA NPs assisted amplification.
Collapse
Affiliation(s)
- Guofu Liao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Xiaofeng Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Xiaohai Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Qing Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China.
| | - Xiuhua Geng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Liyuan Zou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yaqin Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Shaoyuan Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yan Zheng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China.
| |
Collapse
|
73
|
Lorencova L, Bertok T, Bertokova A, Gajdosova V, Hroncekova S, Vikartovska A, Kasak P, Tkac J. Exosomes as a Source of Cancer Biomarkers: Advances in Electrochemical Biosensing of Exosomes. ChemElectroChem 2020. [DOI: 10.1002/celc.202000075] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Lenka Lorencova
- Department of Glycobiotechnology Institute of ChemistrySlovak Academy of Sciences Dubravska cesta 9 845 38 Bratislava Slovakia
| | - Tomas Bertok
- Department of Glycobiotechnology Institute of ChemistrySlovak Academy of Sciences Dubravska cesta 9 845 38 Bratislava Slovakia
| | - Aniko Bertokova
- Department of Glycobiotechnology Institute of ChemistrySlovak Academy of Sciences Dubravska cesta 9 845 38 Bratislava Slovakia
| | - Veronika Gajdosova
- Department of Glycobiotechnology Institute of ChemistrySlovak Academy of Sciences Dubravska cesta 9 845 38 Bratislava Slovakia
| | - Stefania Hroncekova
- Department of Glycobiotechnology Institute of ChemistrySlovak Academy of Sciences Dubravska cesta 9 845 38 Bratislava Slovakia
| | - Alica Vikartovska
- Department of Glycobiotechnology Institute of ChemistrySlovak Academy of Sciences Dubravska cesta 9 845 38 Bratislava Slovakia
| | - Peter Kasak
- Center for Advanced MaterialsQatar University P.O. Box 2713 Doha Qatar
| | - Jan Tkac
- Department of Glycobiotechnology Institute of ChemistrySlovak Academy of Sciences Dubravska cesta 9 845 38 Bratislava Slovakia
| |
Collapse
|
74
|
Freitas M, Nouws HPA, Keating E, Fernandes VC, Delerue-Matos C. Immunomagnetic bead-based bioassay for the voltammetric analysis of the breast cancer biomarker HER2-ECD and tumour cells using quantum dots as detection labels. Mikrochim Acta 2020; 187:184. [PMID: 32088788 DOI: 10.1007/s00604-020-4156-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/11/2020] [Indexed: 12/31/2022]
Abstract
An electrochemical magnetic immunosensing strategy was developed for the determination of HER2-ECD, a breast cancer biomarker, and breast cancer cells in human serum. A sandwich assay was performed on carboxylic acid-functionalized magnetic beads (MBs) using a screen-printed carbon electrode (SPCE) as transducer surface. The affinity process was detected using electroactive labels; core/shell streptavidin-modified CdSe@ZnS Quantum Dots (QDs). Cd2+ ions, released from the QDs, were determined by differential pulse anodic stripping voltammetry (DPASV). An assay time of 90 min, with an actual hands-on time of about 20 min, a linear range between 0.50-50 ng·mL-1 of HER2-ECD and a limit of detection of 0.29 ng·mL-1 were achieved. Analysis of live breast cancer cells was also performed using the optimized assay. Breast cancer cell lines SK-BR-3 (a HER2-positive cell line), MDA-MB-231 (a HER2-negative cell line) and MCF-7 (a cell line with low HER2 expression) were tested. The selectivity of the assay towards SK-BR-3 cells was confirmed. A concentration-dependent signal that was 12.5× higher than the signal obtained for the HER2-negative cells (MDA-MB-231) and a limit of detection of 2 cells·mL-1 was obtained. Graphical abstractSchematic representation of the electrochemical immunomagnetic assay for the determination of the breast cancer biomarker HER2-ECD and cancer cells using magnetic beads (MBs), a screen-printed carbon electrode (SPCE) as transducer surface and quantum dots (QD) as electroactive labels.
Collapse
Affiliation(s)
- Maria Freitas
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, R. Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal
| | - Henri P A Nouws
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, R. Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal.
| | - Elisa Keating
- Department of Biomedicine - Unit of Biochemistry, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.,CINTESIS - Center for Health Technology and Services Research, Rua Dr. Plácido da Costa, 4200-450, Porto, Portugal
| | - Virginia Cruz Fernandes
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, R. Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal
| | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, R. Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal
| |
Collapse
|
75
|
Xia Y, Chen T, Chen G, Weng Y, Zeng L, Liao Y, Chen W, Lan J, Zhang J, Chen J. A nature-inspired colorimetric and fluorescent dual-modal biosensor for exosomes detection. Talanta 2020; 214:120851. [PMID: 32278412 DOI: 10.1016/j.talanta.2020.120851] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/13/2020] [Accepted: 02/15/2020] [Indexed: 12/13/2022]
Abstract
As non-invasive biomarkers, exosomes are of great significance to diseases diagnosis. However, sensitive and accurate detection of exosomes still remains technical challenges. Herein, inspired by nature's "one-to-many" concept, we design a biosensor mimicking the cactus with numerous thorns to detect exosomes. The biosensor is composed of CD63 antibodies, resembling the roots of cactus, to capture exosomes, and the exosomes resemble the stems. Cholesterol-labeled DNA (DNA anchor) binding to streptavidin modified horseradish peroxidase (HRP) can insert into exosomes membrane, which seems the thorns. The readout signal is produced through HRP-catalyzed hydrogen peroxide (H2O2) mediated oxidation of 1,4-phenylenediamine (PPD) to form 2,5-diamino-NN'-bis-(p-aminophenyl)-1,4-benzoquinone di-imine (PPDox). The PPDox can quench fluorescence of fluorescein through inner filter effect (IFE), which provides fluorescent signal for exosomes detection. Based on this principle, the obtained exosomes solution is qualitatively and quantitatively analyzed by our biosensor, with the comparison to current standard methods by nanoparticle tracking analysis (NTA) and commercial enzyme-linked immunosorbent assay (ELISA) kit. The linear range is from 1.0 × 104 to 5.0 × 105 particles μL-1 with the limit of detection 3.40 × 103 particles μL-1 and 3.12 × 103 particles μL-1 for colorimetric and fluorescent assays, respectively. Meanwhile, our biosensor exhibits good selectivity, and can eliminate the interference from proteins. This dual-modal biosensor shows favorable performance towards analytical application in clinic samples, pushing one step further towards practical clinical use.
Collapse
Affiliation(s)
- Yaokun Xia
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China
| | - Tingting Chen
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China
| | - Guanyu Chen
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China
| | - Yunping Weng
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China
| | - Lupeng Zeng
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China
| | - Yijuan Liao
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, 350002, PR China
| | - Wenqian Chen
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China
| | - Jianming Lan
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China
| | - Jing Zhang
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian Province, 350002, PR China.
| | - Jinghua Chen
- Department of Pharmaceutical Analysis, The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China.
| |
Collapse
|
76
|
|
77
|
Freitas M, Neves MMPS, Nouws HPA, Delerue-Matos C. Quantum dots as nanolabels for breast cancer biomarker HER2-ECD analysis in human serum. Talanta 2020; 208:120430. [PMID: 31816682 DOI: 10.1016/j.talanta.2019.120430] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/24/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
Early detection of cancer increases the possibility for an adequate and successful treatment of the disease. Therefore, in this work, a disposable electrochemical immunosensor for the front-line detection of the ExtraCellular Domain of the Human Epidermal growth factor Receptor 2 (HER2-ECD), a breast cancer biomarker, in a simple and efficient manner is presented. Bare screen-printed carbon electrodes were selected as the transducer onto which a sandwich immunoassay was developed. The affinity process was detected through the use of an electroactive label, core/shell CdSe@ZnS Quantum Dots, by differential pulse anodic stripping voltammetry in a total time assay of 2 h, with an actual hands-on time of less than 30 min. The proposed immunosensor responded linearly to HER2-ECD concentration within a wide range (10-150 ng/mL), showing acceptable precision and a limit of detection (2.1 ng/mL, corresponding to a detected amount (sample volume = 40 μL) of 1.18 fmol) which is about 7 times lower than the established cut-off value (15 ng/mL). The usefulness of the developed methodology was tested through the analysis of spiked human serum samples. The reliability of the presented biosensor for the selective screening of HER2-ECD was confirmed by analysing another breast cancer biomarker (CA15-3) and several human serum proteins.
Collapse
Affiliation(s)
- Maria Freitas
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal
| | - Marta M P S Neves
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal
| | - Henri P A Nouws
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal.
| | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Politécnico do Porto, Rua Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal
| |
Collapse
|
78
|
Gaillard M, Thuaire A, Nonglaton G, Agache V, Roupioz Y, Raillon C. Biosensing extracellular vesicles: contribution of biomolecules in affinity-based methods for detection and isolation. Analyst 2020; 145:1997-2013. [PMID: 31960838 DOI: 10.1039/c9an01949a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular Vesicles (EVs) are lipid vesicles secreted by cells that allow intercellular communication. They are decorated with surface proteins, which are membrane proteins that can be targeted by biochemical techniques to isolate EVs from background particles. EVs have recently attracted attention for their potential applications as biomarkers for numerous diseases. This review focuses on the contribution of biomolecules used as ligands in affinity-based biosensors for the detection and isolation of EVs. Capturing biological objects like EVs with antibodies is well described in the literature through different biosensing techniques. However, since handling proteins can be challenging due to stability issues, sensors using non-denaturable biomolecules are emerging. DNA aptamers, short DNA fragments that mimic antibody action, are currently being developed and considered as the future of antibody-like ligands. These molecules offer undeniable advantages: unparalleled ease of production, very high stability in air, similar affinity constants to antibodies, and compatibility with many organic solvents. The use of peptides specific to EVs is also an exciting biochemical solution to target EV membrane proteins and complement other probes. These different ligands have been used in several types of biosensors: electrochemical, optical, microfluidic using both generic probes (targeting widely expressed membrane proteins such as the tetraspanins) and specific probes (targeting disease biomarkers such as proteins overexpressed in cancer).
Collapse
Affiliation(s)
- M Gaillard
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, 38000 Grenoble, France.
| | | | | | | | | | | |
Collapse
|
79
|
Chen Z, Yamaguchi H, Kameoka J. Development of size-selective microfluidic platform. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:5661-5664. [PMID: 31947137 DOI: 10.1109/embc.2019.8857536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Exosomes are nanosized extracellular vesicles that play a significant role in cell-cell communication. Recently, there is significant interest in exosome-related fundamental research, especially subgroups of exosomes as potential biomarkers for cancer diagnosis and prognosis. In this paper, we report a new size selective isolation method via elastic lift force and nanomembrane filtration and demonstrated the liposome recovery rate of 92.5% from a mixture solution of 1 μm polystyrene beads, 100 nm liposomes and proteins as a proof of concept for exosome isolation. This single microfluidic platform offers an improved approach with short processing time (<; 2 hours) and low cost, and shows potential broad applicability to cancer biomarker studies.
Collapse
|
80
|
Martín-Gracia B, Martín-Barreiro A, Cuestas-Ayllón C, Grazú V, Line A, Llorente A, M. de la Fuente J, Moros M. Nanoparticle-based biosensors for detection of extracellular vesicles in liquid biopsies. J Mater Chem B 2020; 8:6710-6738. [DOI: 10.1039/d0tb00861c] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Selecting the appropriate nanoparticle, functionalization chemistry and sensing methodology can speed up the translation of liquid biopsies into the clinic.
Collapse
Affiliation(s)
- Beatriz Martín-Gracia
- Aragón Materials Science Institute (ICMA)
- CSIC/University of Zaragoza
- Zaragoza
- Spain
- Biomedical Research Networking Center in Bioengineering
| | - Alba Martín-Barreiro
- Aragón Materials Science Institute (ICMA)
- CSIC/University of Zaragoza
- Zaragoza
- Spain
- Biomedical Research Networking Center in Bioengineering
| | | | - Valeria Grazú
- Aragón Materials Science Institute (ICMA)
- CSIC/University of Zaragoza
- Zaragoza
- Spain
- Biomedical Research Networking Center in Bioengineering
| | - Aija Line
- Latvian Biomedical Research and Study Centre
- Riga
- Latvia
| | - Alicia Llorente
- Department of Molecular Cell Biology
- Institute for Cancer Research
- Oslo University Hospital
- Oslo
- Norway
| | - Jesús M. de la Fuente
- Aragón Materials Science Institute (ICMA)
- CSIC/University of Zaragoza
- Zaragoza
- Spain
- Biomedical Research Networking Center in Bioengineering
| | - María Moros
- Aragón Materials Science Institute (ICMA)
- CSIC/University of Zaragoza
- Zaragoza
- Spain
- Biomedical Research Networking Center in Bioengineering
| |
Collapse
|
81
|
Akkilic N, Geschwindner S, Höök F. Single-molecule biosensors: Recent advances and applications. Biosens Bioelectron 2019; 151:111944. [PMID: 31999573 DOI: 10.1016/j.bios.2019.111944] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023]
Abstract
Single-molecule biosensors serve the unmet need for real time detection of individual biological molecules in the molecular crowd with high specificity and accuracy, uncovering unique properties of individual molecules which are hidden when measured using ensemble averaging methods. Measuring a signal generated by an individual molecule or its interaction with biological partners is not only crucial for early diagnosis of various diseases such as cancer and to follow medical treatments but also offers a great potential for future point-of-care devices and personalized medicine. This review summarizes and discusses recent advances in nanosensors for both in vitro and in vivo detection of biological molecules offering single-molecule sensitivity. In the first part, we focus on label-free platforms, including electrochemical, plasmonic, SERS-based and spectroelectrochemical biosensors. We review fluorescent single-molecule biosensors in the second part, highlighting nanoparticle-amplified assays, digital platforms and the utilization of CRISPR technology. We finally discuss recent advances in the emerging nanosensor technology of important biological species as well as future perspectives of these sensors.
Collapse
Affiliation(s)
- Namik Akkilic
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Stefan Geschwindner
- Structure, Biophysics and Fragment-based Lead Generation, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Fredrik Höök
- Department of Applied Physics, Division of Biological Physics, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
82
|
Li W, Wang H, Zhao Z, Gao H, Liu C, Zhu L, Wang C, Yang Y. Emerging Nanotechnologies for Liquid Biopsy: The Detection of Circulating Tumor Cells and Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805344. [PMID: 30589111 DOI: 10.1002/adma.201805344] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/29/2018] [Indexed: 05/18/2023]
Abstract
Liquid biopsy enables noninvasive and dynamic analysis of molecular or cellular biomarkers, and therefore holds great potential for the diagnosis, prognosis, monitoring of disease progress and treatment efficacy, understanding of disease mechanisms, and identification of therapeutic targets for drug development. In this review, the recent progress in nanomaterials, nanostructures, nanodevices, and nanosensors for liquid biopsy is summarized, with a focus on the detection and molecular characterization of circulating tumor cells (CTCs) and extracellular vesicles (EVs). The developments and advances of nanomaterials and nanostructures in enhancing the sensitivity, specificity, and purity for the detection of CTCs and EVs are discussed. Sensing techniques for signal transduction and amplification as well as visualization strategies are also discussed. New technologies for the reversible release of the isolated CTCs and EVs and for single-CTC/EV analysis are summarized. Emerging microfluidic platforms for the integral on-chip isolation, detection, and molecular analysis are also included. The opportunities, challenges, and prospects of these innovative materials and technologies, especially with regard to their feasibility in clinical applications, are discussed. The applications of nanotechnology-based liquid biopsy will bring new insight into the clinical practice in monitoring and treatment of tumor and other significant diseases.
Collapse
Affiliation(s)
- Wenzhe Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huayi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zijian Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Houqian Gao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Changliang Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
83
|
Cheng N, Du D, Wang X, Liu D, Xu W, Luo Y, Lin Y. Recent Advances in Biosensors for Detecting Cancer-Derived Exosomes. Trends Biotechnol 2019; 37:1236-1254. [DOI: 10.1016/j.tibtech.2019.04.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/14/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023]
|
84
|
Soda N, Rehm BHA, Sonar P, Nguyen NT, Shiddiky MJA. Advanced liquid biopsy technologies for circulating biomarker detection. J Mater Chem B 2019; 7:6670-6704. [PMID: 31646316 DOI: 10.1039/c9tb01490j] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liquid biopsy is a new diagnostic concept that provides important information for monitoring and identifying tumor genomes in body fluid samples. Detection of tumor origin biomolecules like circulating tumor cells (CTCs), circulating tumor specific nucleic acids (circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA), microRNAs (miRNAs), long non-coding RNAs (lnRNAs)), exosomes, autoantibodies in blood, saliva, stool, urine, etc. enables cancer screening, early stage diagnosis and evaluation of therapy response through minimally invasive means. From reliance on painful and hazardous tissue biopsies or imaging depending on sophisticated equipment, cancer management schemes are witnessing a rapid evolution towards minimally invasive yet highly sensitive liquid biopsy-based tools. Clinical application of liquid biopsy is already paving the way for precision theranostics and personalized medicine. This is achieved especially by enabling repeated sampling, which in turn provides a more comprehensive molecular profile of tumors. On the other hand, integration with novel miniaturized platforms, engineered nanomaterials, as well as electrochemical detection has led to the development of low-cost and simple platforms suited for point-of-care applications. Herein, we provide a comprehensive overview of the biogenesis, significance and potential role of four widely known biomarkers (CTCs, ctDNA, miRNA and exosomes) in cancer diagnostics and therapeutics. Furthermore, we provide a detailed discussion of the inherent biological and technical challenges associated with currently available methods and the possible pathways to overcome these challenges. The recent advances in the application of a wide range of nanomaterials in detecting these biomarkers are also highlighted.
Collapse
Affiliation(s)
- Narshone Soda
- School of Environment and Science, Griffith University, Nathan Campus, QLD 4111, Australia. and Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, QLD 4111, Australia
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers (CCFB), Griffith Institute for Drug Discovery (GRIDD), Griffith University, Nathan, QLD 4111, Australia
| | - Prashant Sonar
- School of Chemistry, Physics and Mechanical Engineering, Molecular Design and Synthesis, Queensland University of Technology (QUT), Brisbane, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, QLD 4111, Australia
| | - Muhammad J A Shiddiky
- School of Environment and Science, Griffith University, Nathan Campus, QLD 4111, Australia. and Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, QLD 4111, Australia
| |
Collapse
|
85
|
Lv X, Geng Z, Su Y, Fan Z, Wang S, Fang W, Chen H. Label-Free Exosome Detection Based on a Low-Cost Plasmonic Biosensor Array Integrated with Microfluidics. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:9816-9824. [PMID: 31268344 DOI: 10.1021/acs.langmuir.9b01237] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Localized surface plasmon resonance-based plasmonic biosensors are interesting candidates for the design of portable optical biosensor platforms owing to their integration, miniaturization, multiparameter, real-time, and label-free detection characteristics. Plasmonic biosensor arrays that have been combined with microfluidics have been developed herein to detect exosomes label-free. Gold nano-ellipsoid arrays were fabricated with low-cost anodic aluminum oxide thin films that act as shadow masks for evaporation of Au. The nano-ellipsoid arrays were integrated with a microfluidic chip to achieve multiparameter detection. The anti-CD63 antibody that is specific to the exosome transmembrane protein CD63 is modified on the surface of the nano-ellipsoids. Exosome samples were injected into the biosensor platform at different concentrations and detected successfully. The detection limit was 1 ng/mL. The proposed plasmonic biosensor array can be universally applicable for the detection of other biomarkers by simply changing the antibody on the surface of the Au nano-ellipsoids. Moreover, this biosensor platform is envisaged to be potentially useful in the development of low-cost plasmonic-based biosensors for biomarker detection and for the investigation of exosomes for noninvasive disease diagnoses.
Collapse
Affiliation(s)
- Xiaoqing Lv
- State Key Laboratory of Integrated Opoelectronics, Institute of Semiconductors , Chinese Academy of Sciences , Beijing 100083 , China
| | - Zhaoxin Geng
- State Key Laboratory of Integrated Opoelectronics, Institute of Semiconductors , Chinese Academy of Sciences , Beijing 100083 , China
- School of Information Engineering , Minzu University of China , Beijing 100081 , China
| | - Yue Su
- State Key Laboratory of Integrated Opoelectronics, Institute of Semiconductors , Chinese Academy of Sciences , Beijing 100083 , China
- College of Materials Science and Opto-Electronic Technology , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Zhiyuan Fan
- State Key Laboratory of Integrated Opoelectronics, Institute of Semiconductors , Chinese Academy of Sciences , Beijing 100083 , China
- College of Materials Science and Opto-Electronic Technology , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Shicai Wang
- State Key Laboratory of Integrated Opoelectronics, Institute of Semiconductors , Chinese Academy of Sciences , Beijing 100083 , China
- State Key Laboratory of Crystal Materials , Shandong University , Jinan 250022 , China
| | - Weihao Fang
- State Key Laboratory of Integrated Opoelectronics, Institute of Semiconductors , Chinese Academy of Sciences , Beijing 100083 , China
- College of Materials Science and Opto-Electronic Technology , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Hongda Chen
- State Key Laboratory of Integrated Opoelectronics, Institute of Semiconductors , Chinese Academy of Sciences , Beijing 100083 , China
| |
Collapse
|
86
|
Yu Y, Li YT, Jin D, Yang F, Wu D, Xiao MM, Zhang H, Zhang ZY, Zhang GJ. Electrical and Label-Free Quantification of Exosomes with a Reduced Graphene Oxide Field Effect Transistor Biosensor. Anal Chem 2019; 91:10679-10686. [PMID: 31331170 DOI: 10.1021/acs.analchem.9b01950] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yi Yu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan 430065, People’s Republic of China
| | - Yu-Tao Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan 430065, People’s Republic of China
| | - Dan Jin
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan 430065, People’s Republic of China
| | - Fan Yang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan 430065, People’s Republic of China
| | - Ding Wu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan 430065, People’s Republic of China
| | - Meng-Meng Xiao
- Key Laboratory for the Physics and Chemistry of Nanodevices, Department of Electronics, Peking University, 5 Yiheyuan Road, Beijing 100871, People’s Republic of China
| | - Hong Zhang
- Teaching and Research Office of Forensic Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan 430065, People’s Republic of China
| | - Zhi-Yong Zhang
- Key Laboratory for the Physics and Chemistry of Nanodevices, Department of Electronics, Peking University, 5 Yiheyuan Road, Beijing 100871, People’s Republic of China
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 1 Huangjia Lake West Road, Wuhan 430065, People’s Republic of China
| |
Collapse
|
87
|
Groza M, Zimta AA, Irimie A, Achimas-Cadariu P, Cenariu D, Stanta G, Berindan-Neagoe I. Recent advancements in the study of breast cancer exosomes as mediators of intratumoral communication. J Cell Physiol 2019; 235:691-705. [PMID: 31328284 DOI: 10.1002/jcp.29096] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/25/2019] [Indexed: 12/24/2022]
Abstract
Breast cancer is a heterogeneous disease, with a morbidity rate of 27.8% and a mortality rate of 15% among women population worldwide. Understanding how this cancer develops and the mechanisms behind tumor progression and chemoresistance is of utmost importance. Exosomes mediate communication in a population of heterogeneous tumoral cells. They have a cargo composed of oncogenes and oncomiRs which change the transcriptomic scenario of their targeted cells and activate numerous tumor-promoting signaling pathways. Exosomes secreted by breast cancer cells lead to enhanced cell proliferation, replicative immortality, angiogenesis, invasion, migration, and chemoresistance. Studying exosomes from this perspective offers more in depth understanding of breast malignancy and may aid in the future development of early diagnostic, prognostic and therapeutic options. We present the latest findings in this area and offer practical solutions which may further stimulate the much-needed research of exosome in breast cancer.
Collapse
Affiliation(s)
- Monica Groza
- Department of Medical Oncology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, Cluj-Napoca, Romania
| | - Alexandru Irimie
- 11th Department of Oncological Surgery and Gynecological Oncology, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania.,Department of Surgery, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Patriciu Achimas-Cadariu
- 11th Department of Oncological Surgery and Gynecological Oncology, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania.,Department of Surgery, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Diana Cenariu
- MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, Cluj-Napoca, Romania
| | - Giorgio Stanta
- DSM, Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Ioana Berindan-Neagoe
- MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, Cluj-Napoca, Romania.,Research Center for Functional Genomics, Biomedicine and Translational Medicine, uliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| |
Collapse
|
88
|
An Y, Jin T, Zhu Y, Zhang F, He P. An ultrasensitive electrochemical aptasensor for the determination of tumor exosomes based on click chemistry. Biosens Bioelectron 2019; 142:111503. [PMID: 31376716 DOI: 10.1016/j.bios.2019.111503] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/19/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022]
Abstract
Exosomes, lipid bilayer membrane vesicles, can guide various pathological and physiological processes. However, reliable, convenient and sensitive methods for exosome determination for early cancer diagnosis are still technically challenging. Herein, an electrochemical aptasensor based on click chemistry and the DNA hybridization chain reaction (HCR) for signal amplification has been developed for the ultrasensitive detection of tumor exosomes. CD63 aptamer was first immobilized on a glassy carbon electrode for capturing exosomes, and 4-oxo-2-nonenal alkyne (alkynyl-4-ONE) molecules, functionalized lipid electrophiles, were conjugated to the exosomes via the reaction of amino and aldehyde groups. Azide-labeled DNA probe as an anchor was then connected to the exosomes by copper (I)-catalyzed click chemistry. Signal amplification was achieved by HCR, and the numerous linked horseradish peroxidase (HRP) molecules could catalyze the reaction of o-phenylenediamine (OPD) and H2O2. The concentration of exosomes could be quantified by monitoring the electrochemical reduction current of 2,3-diaminophenazine (DAP). Under the optimal conditions, this method allowed the sensitive detection of exosomes in the range of 1.12 × 102 to 1.12 × 108 particles/μL with a limit of detection (LOD) of 96 particles/μL. Furthermore, the present assay enabled sensitive and accurate quantification of exosomes in human serum, and it has high potential for exosome analysis in clinical samples.
Collapse
Affiliation(s)
- Yu An
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Tongyu Jin
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Yuyuan Zhu
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| | - Fan Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China.
| | - Pingang He
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, PR China
| |
Collapse
|
89
|
Zhang Z, Tang C, Zhao L, Xu L, Zhou W, Dong Z, Yang Y, Xie Q, Fang X. Aptamer-based fluorescence polarization assay for separation-free exosome quantification. NANOSCALE 2019; 11:10106-10113. [PMID: 31089660 DOI: 10.1039/c9nr01589b] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Tumor-derived exosomes have emerged as promising cancer biomarkers and attracted increasing interest in non-invasive cancer diagnosis and treatment monitoring. However, the identification and quantification of exosomes in clinical samples such as blood remains challenging due to the difficulty in trade-off between recognition specificity and isolation efficiency. Here we have developed an aptamer-based fluorescence polarization assay for exosome quantification, which is a separation-free, amplification-free and sensitive approach enabling direct quantification of exosomes in human plasma. While the key specificity of this assay is based on the aptamer's inherent affinity to membrane proteins on exosomes, exosomes' inherent huge mass/volume acts as mass-based fluorescence polarization amplifier. Our assay allows quantitative analysis of exosomes in the range of 5 × 102 to 5 × 105 particles per μL with a detect limitation of 500 particles per μL for the cell line deprived exosomes. The total assay time is about 30 min with just one mix-and-read step to achieve high sensitivity. We have also demonstrated quantification of exosomes from lung cancer patients and healthy donors in clinical samples. This work describes a new and simple liquid biopsy assay to directly detect exosomes in the biological matrix, which facilitates cancer diagnosis and therapy monitoring.
Collapse
Affiliation(s)
- Zhen Zhang
- CAS Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing National Research Center for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Chuanhao Tang
- Department of Medical Oncology, Peking University International Hospital, Beijing 102206, P. R. China
| | - Libo Zhao
- Echo Biotech Co. Ltd, Beijing 102206, P. R. China
| | - Li Xu
- CAS Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing National Research Center for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Wei Zhou
- CAS Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing National Research Center for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zaizai Dong
- CAS Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing National Research Center for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yuqing Yang
- Echo Biotech Co. Ltd, Beijing 102206, P. R. China
| | - Qiqi Xie
- Echo Biotech Co. Ltd, Beijing 102206, P. R. China
| | - Xiaohong Fang
- CAS Key Laboratory of Molecular Nanostructure and Nanotechnology, Beijing National Research Center for Molecular Sciences, CAS Research/Education Center for Excellence in Molecule Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
90
|
Qiao B, Guo Q, Jiang J, Qi Y, Zhang H, He B, Cai C, Shen J. An electrochemiluminescent aptasensor for amplified detection of exosomes from breast tumor cells (MCF-7 cells) based on G-quadruplex/hemin DNAzymes. Analyst 2019; 144:3668-3675. [PMID: 31086892 DOI: 10.1039/c9an00181f] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exosomes are non-invasive biomarkers for cancer diagnosis. Herein, we describe an electrochemiluminescent (ECL) aptasensor for the detection of exosomes from breast tumor cells. Mercaptopropionic acid (MPA)-modified Eu3+-doped CdS nanocrystals (MPA-CdS:Eu NCs) and H2O2 were used as ECL emitters and coreactant, respectively. The exosomes are recognized and captured by the CD63 aptamer, and then form a G-quadruplex/hemin DNAzyme, which efficiently catalyzes the decomposition of H2O2, resulting in the decreased ECL signal of MPA-CdS:Eu NCs. The exosomes from breast tumor cells (MCF-7 cells) can be detected in the range of 3.4 × 105 to 1.7 × 108 particles per mL. The limit of detection (LOD) was estimated to be 7.41 × 104 particles per mL at a signal-to-noise ratio of 3. The aptasensor has been successfully used to detect exosomes in the serum.
Collapse
Affiliation(s)
- Bin Qiao
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Qunqun Guo
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Juqian Jiang
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Yunlong Qi
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Hui Zhang
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Bangshun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, P. R. China.
| | - Chenxin Cai
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| | - Jian Shen
- Jiangsu Key Laboratory of Biomedical Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, National and Local Joint Engineering Research Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, P. R. China.
| |
Collapse
|
91
|
Pastucha M, Farka Z, Lacina K, Mikušová Z, Skládal P. Magnetic nanoparticles for smart electrochemical immunoassays: a review on recent developments. Mikrochim Acta 2019; 186:312. [PMID: 31037494 DOI: 10.1007/s00604-019-3410-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/03/2019] [Indexed: 02/07/2023]
Abstract
This review (with 129 refs) summarizes the progress in electrochemical immunoassays combined with magnetic particles that was made in the past 5 years. The specifity of antibodies linked to electrochemical transduction (by amperometry, voltammetry, impedimetry or electrochemiluminescence) gains further attractive features by introducing magnetic nanoparticles (MNPs). This enables fairly easy preconcentration of analytes, minimizes matrix effects, and introduces an appropriate label. Following an introduction into the fundamentals of electrochemical immunoassays and on nanomaterials for respective uses, a large chapter addresses method for magnetic capture and preconcentration of analytes. A next chapter discusses commonly used labels such as dots, enzymes, metal and metal oxide nanoparticles and combined clusters. The large field of hybrid nanomaterials for use in such immunoassays is discussed next, with a focus on MNPs composites with various kinds of graphene variants, polydopamine, noble metal nanoparticles or nanotubes. Typical applications address clinical markers (mainly blood and urine parameters), diagnosis of cancer (markers and cells), detection of pathogens (with subsections on viruses and bacteria), and environmental and food contaminants as toxic agents and pesticides. A concluding section summarizes the present status, current challenges, and highlights future trends. Graphical abstract Magnetic nanoparticles (MNP) with antibodies (Ab) capture and preconcentrate analyte from sample (a) and afterwards become magnetically (b) or immunospecifically (c) bound at an electrode. Signal either increases due to the presence of alabel (b) or decreases as the redox probe is blocked (c).
Collapse
Affiliation(s)
- Matěj Pastucha
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Zdeněk Farka
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Karel Lacina
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Zuzana Mikušová
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Skládal
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| |
Collapse
|
92
|
Laopa P, Vilaivan T. Cationic‐Polymer‐Functionalized Zinc Oxide Quantum Dots: Preparation and Application to Iron(II) Ion Detection. ChemistrySelect 2019. [DOI: 10.1002/slct.201900424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Praethong Laopa
- Department of General Science, Faculty of Science and EngineeringKasetsart UniversityChalermphrakiat Sakon Nakhon Province Campus Sakon Nakhon 47000 Thailand
| | - Tirayut Vilaivan
- Organic Synthesis Research UnitDepartment of Chemistry, Faculty of ScienceChulalongkorn University Phayathai Road, Patumwan Bangkok 10330 Thailand
| |
Collapse
|
93
|
Boriachek K, Masud MK, Palma C, Phan HP, Yamauchi Y, Hossain MSA, Nguyen NT, Salomon C, Shiddiky MJA. Avoiding Pre-Isolation Step in Exosome Analysis: Direct Isolation and Sensitive Detection of Exosomes Using Gold-Loaded Nanoporous Ferric Oxide Nanozymes. Anal Chem 2019; 91:3827-3834. [DOI: 10.1021/acs.analchem.8b03619] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kseniia Boriachek
- School of Environment and Science, Griffith University Nathan Campus, Nathan, Queensland 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University Nathan Campus, Nathan, Queensland 4111, Australia
| | - Mostafa Kamal Masud
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University Nathan Campus, Nathan, Queensland 4111, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Carlos Palma
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane, Queensland 4029, Australia
| | - Hoang-Phuong Phan
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University Nathan Campus, Nathan, Queensland 4111, Australia
| | - Yusuke Yamauchi
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Chemical Engineering, Faculty of Engineering, Architecture and Information Technology (EAIT), The University of Queensland, Brisbane, Queensland 4072, Australia
- Department of Plant & Environmental New Resources, Kyung Hee University, 1732 Deogyeong-daero, Giheunggu, Yongin-si, Gyeonggi-do 446-701, South Korea
| | - Md. Shahriar A. Hossain
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, Queensland 4072, Australia
- School of Mechanical & Mining Engineering, Faculty of Engineering, Architecture and Information Technology (EAIT), The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nam-Trung Nguyen
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University Nathan Campus, Nathan, Queensland 4111, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane, Queensland 4029, Australia
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción 4030000, Chile
| | - Muhammad J. A. Shiddiky
- School of Environment and Science, Griffith University Nathan Campus, Nathan, Queensland 4111, Australia
- Queensland Micro and Nanotechnology Centre (QMNC), Griffith University Nathan Campus, Nathan, Queensland 4111, Australia
| |
Collapse
|
94
|
Wongkaew N. Nanofiber-integrated miniaturized systems: an intelligent platform for cancer diagnosis. Anal Bioanal Chem 2019; 411:4251-4264. [PMID: 30706075 DOI: 10.1007/s00216-019-01589-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/26/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022]
Abstract
Cancer diagnostic tools enabling screening, diagnosis, and effective disease management are essential elements to increase the survival rate of diagnosed patients. Low abundance of cancer markers present in large amounts of interferences remains the major issue. Moreover, current diagnostic technologies are restricted to high-resourced settings only. Integrating nanofibers into miniaturized analytical systems holds a significant promise to address these challenges as demonstrated by recent publications. A large surface area, three-dimensional porous network, and diverse range of functional chemistries make nanofibers an excellent candidate as immobilization support and/or transduction elements, enabling high capture yield and ultrasensitive detection in miniaturized devices. Functional nanofibers have thus been used to isolate and detect various cancer-related biomarkers with a high degree of success in both on-chip and off-chip platforms. In fact, the chemical and functional adaptability of nanofibers has been exploited to address the technical challenges unique to each of the cancer markers in body fluids, where circulating tumor cells are prominently investigated among others (proteins, nucleic acids, and exosomes). So far, none of the work has exploited the nanofibers for cancer-derived exosomes, opening an avenue for further research effort. The trend and future prospects signal possibilities to strengthen the implementation of nanofiber-miniaturized system hybrid for a next generation of cancer diagnostic platforms both in clinical and point-of-care testing.
Collapse
Affiliation(s)
- Nongnoot Wongkaew
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany.
| |
Collapse
|
95
|
Cheshomi H, Matin MM. Exosomes and their importance in metastasis, diagnosis, and therapy of colorectal cancer. J Cell Biochem 2019; 120:2671-2686. [PMID: 30246315 DOI: 10.1002/jcb.27582] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/07/2018] [Indexed: 01/24/2023]
Abstract
Extracellular vesicles are known as actual intermediaries of intercellular communications, such as biological signals and cargo transfer between different cells. A variety of cells release the exosomes as nanovesicular bodies. Exosomes contain different compounds such as several types of nucleic acids and proteins. In this study, we focused on exosomes in colorectal cancer as good tools that can be involved in various cancer-related processes. Furthermore, we summarize the advantages and disadvantages of exosome extraction methods and review related studies on the role of exosomes in colorectal cancer. Finally, we focus on reports available on relations between mesenchymal stem cell-derived exosomes and colorectal cancer. Several cancer-related processes such as cancer progression, metastasis, and drug resistance of colorectal cancer are related to the cargoes of exosomes. A variety of molecules, especially proteins, microRNAs, and long noncoding RNAs, play important roles in these processes. The microenvironment features, such as hypoxia, also have very important effects on the properties of the origin cell-derived exosomes. On the other hand, exosomes derived from colorectal cancer cells also interfere with cancer chemoresistance. Furthermore, today it is known that exosomes and their contents can likely be very effective in noninvasive colorectal cancer diagnosis and therapy. Thus, exosomes, and especially their cargoes, play different key roles in various aspects of basic and clinical research related to both progression and therapy of colorectal cancer.
Collapse
Affiliation(s)
- Hamid Cheshomi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
96
|
Hartjes TA, Mytnyk S, Jenster GW, van Steijn V, van Royen ME. Extracellular Vesicle Quantification and Characterization: Common Methods and Emerging Approaches. Bioengineering (Basel) 2019; 6:bioengineering6010007. [PMID: 30654439 PMCID: PMC6466085 DOI: 10.3390/bioengineering6010007] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are a family of small membrane vesicles that carry information about cells by which they are secreted. Growing interest in the role of EVs in intercellular communication, but also in using their diagnostic, prognostic and therapeutic potential in (bio) medical applications, demands for accurate assessment of their biochemical and physical properties. In this review, we provide an overview of available technologies for EV analysis by describing their working principles, assessing their utility in EV research and summarising their potential and limitations. To emphasise the innovations in EV analysis, we also highlight the unique possibilities of emerging technologies with high potential for further development.
Collapse
Affiliation(s)
- Thomas A Hartjes
- Department of Pathology, Erasmus Optical Imaging Centre, Erasmus MC, 3015 GE Rotterdam, The Netherlands.
| | - Serhii Mytnyk
- Department of Chemical Engineering, Delft University of Technology, 3015 CD Delft, The Netherlands.
| | - Guido W Jenster
- Department of Urology, Erasmus MC, 3015 CD Rotterdam, The Netherlands.
| | - Volkert van Steijn
- Department of Chemical Engineering, Delft University of Technology, 3015 CD Delft, The Netherlands.
| | - Martin E van Royen
- Department of Pathology, Erasmus Optical Imaging Centre, Erasmus MC, 3015 GE Rotterdam, The Netherlands.
| |
Collapse
|
97
|
Zhou YG, Kermansha L, Zhang L, Mohamadi RM. Miniaturized Electrochemical Sensors to Facilitate Liquid Biopsy for Detection of Circulating Tumor Markers. Bioanalysis 2019. [DOI: 10.1007/978-981-13-6229-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
98
|
Yang B, Chen Y, Shi J. Exosome Biochemistry and Advanced Nanotechnology for Next-Generation Theranostic Platforms. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1802896. [PMID: 30126052 DOI: 10.1002/adma.201802896] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/07/2018] [Indexed: 05/26/2023]
Abstract
Recent marked technological advances in the field of exosome nanotechnology have provided unprecedented opportunities to bloom the developments of exosome-related biology, chemistry, pathology, and therapeutics, which have laid a solid basis for scientific community to design exosome-based nanotheranostic platforms. The unique structural/compositional/morphological characteristics of exosomes as natural nanocarriers, as well as their fascinating physicochemical/biochemical properties, which underpin their special physiopathological roles, have triggered the concept that these cell-derived nanovesicles with intrinsic biological functions can be highly competent for the establishment of next-generation nanomedicine. Herein, efforts are made to give a comprehensive overview on the recent advances of exosome nanotechnology based on the representative examples of the current state of the art of exosome-based research, ranging from their formation, biological function, preparation, and characterization to their extensive nanomedical applications. It is highly expected that the better and clearer elucidation of the fundamental principles for advanced nanotechnology in constructing exosome-based theranostic nanoplatforms, as well as integrating the intrinsic advantages of exosomes as endogenous cell-derived nanocarriers with the advanced design methodology of traditional nanomedicine, will help to unlock the innate powers of exosomes for the establishment of next-generation theranostic nanoplatforms.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
99
|
Sahoo SL, Liu CH, Kumari M, Wu WC, Wang CC. Biocompatible quantum dot-antibody conjugate for cell imaging, targeting and fluorometric immunoassay: crosslinking, characterization and applications. RSC Adv 2019; 9:32791-32803. [PMID: 35529742 PMCID: PMC9073096 DOI: 10.1039/c9ra07352c] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/01/2019] [Indexed: 11/21/2022] Open
Abstract
Quantum dots (QDs) are important fluorescent probes that offer great promise for bio-imaging research due to their superior optical properties. However, QDs for live cell imaging and the tracking of cells need more investigation to simplify processing procedures, improving labeling efficiency, and reducing chronic toxicity. In this study, QDs were functionalized with bovine serum albumin (BSA) via a chemical linker. Anti-human immunoglobulin antibodies were oxidized by sodium periodate to create reactive aldehyde groups for a spontaneous reaction with the amine groups of BSA-modified QDs. An antibody-labeled QD bioconjugate was characterized using agarose gel electrophoresis, dynamic light scattering, and zeta potential. Using fluorescence spectroscopy, we found that the fluorescence of QDs was retained after multiple conjugation steps. The cell-labeling function of the QD bioconjugate was confirmed using an image analyzer and confocal microscopy. The QD bioconjugate specifically targeted human immunoglobulin on the membrane surface of recombinant cells. In addition, the QD bioconjugate applied in fluorometric immunoassay was effective for the quantitative analysis of human immunoglobulin in an enzyme-linked immunosorbent assay. The developed QD bioconjugate may offer a promising platform to develop biocompatible tools to label cells and quantify antibodies in the immunoassay. A layer-by-layer covalent strategy is developed including the modification of QDs using BSA as a stabilizing agent and then anti-human immunoglobulin antibody as a targeting moiety.![]()
Collapse
Affiliation(s)
- Soubhagya Laxmi Sahoo
- Department of Chemical and Materials Engineering
- Chang Gung University
- Tao-Yuan 333
- Taiwan
| | - Chi-Hsien Liu
- Department of Chemical and Materials Engineering
- Chang Gung University
- Tao-Yuan 333
- Taiwan
- Research Center for Chinese Herbal Medicine
| | - Monika Kumari
- Department of Chemical and Materials Engineering
- Chang Gung University
- Tao-Yuan 333
- Taiwan
| | - Wei-Chi Wu
- Department of Ophthalmology
- Chang Gung Memorial Hospital
- Taoyuan
- Taiwan
- College of Medicine
| | - Chun-Chao Wang
- Department of Medical Science
- Institute of Molecular Medicine
- National Tsing Hua University
- Hsinchu
- Taiwan
| |
Collapse
|
100
|
Zhang LJ, Xia L, Xie HY, Zhang ZL, Pang DW. Quantum Dot Based Biotracking and Biodetection. Anal Chem 2018; 91:532-547. [DOI: 10.1021/acs.analchem.8b04721] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Li-Juan Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
| | - Li Xia
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
| | - Hai-Yan Xie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Zhi-Ling Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
| | - Dai-Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
- College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| |
Collapse
|