1
|
Haizan I, Choi MY, Park DH, Choi JH. Dual-target magneto-immunoassay with bifunctional nanohybrids for breast cancer exosome detection. Talanta 2025; 286:127532. [PMID: 39788070 DOI: 10.1016/j.talanta.2025.127532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Exosomes, crucial for intercellular communication, hold potential as noninvasive liquid biopsy biomarkers especially in early breast cancer detection benefitted from the distinctive "cancer signature" on their membrane surface. Yet, the present methodologies of exosomes for breast cancer detection have involved the implementation of only a single member from the tetraspanin protein group as a biomarker. Moreso, due to the high concentration of exosomes in complex body fluids, there is a compelling need to measure a small concentration of cancer-derived exosomes with a low background noise signal. In this study, we designed and characterized magnetic core-gold shell nanohybrids (mAuNHs) that function as detection and isolator probes, which were integrated in a simple colorimetric sandwich magneto-immunoassay (mLISA). The magnetic core of mAuNHs facilitates the separation of exosomes from complex samples of biological origin whereby amorphous structures were effectively removed, decreasing background signal. Meanwhile, the coalescence effect of pairing biologically abundance exosomal marker (CD9 antibody) with the cancer specific (CD24 antibody) offers a highly selective and sensitive detection of our target model, MCF7 exosomes. As a result, using our mLISA system, exosomes derived from MCF7 can be selectively recognized from other tested cancer cell lines, BT474 and PC3. Besides, as low as 37 particles/μL of limit of detection (LOD) was achieved using mLISA sensor, exhibiting a good sensitivity as compared to conventional ELISA. Overall, our proposed dual-target biosensor offers a great reduction on background noise from samples, simplicity for users as in exosome's lengthy preparation is reduced as well as good sensitivity.
Collapse
Affiliation(s)
- Izzati Haizan
- Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| | - Min Yu Choi
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| | - Dong Hyeok Park
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| | - Jin-Ha Choi
- Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea; School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| |
Collapse
|
2
|
Sulipta JS, Jeong H, Hong S. Polydopamine as a versatile optical indicator for colorimetric and fluorescence-based biosensing. Biomater Sci 2025. [PMID: 40111266 DOI: 10.1039/d5bm00084j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Beyond their well-established adhesive properties, polydopamine (pDA) and pDA-like materials are emerging as superior alternatives to conventional optical indicators in biosensing applications due to their exceptional biocompatibility, tunable optical properties, and high sensitivity, arising from their eumelanin-like physicochemical characteristics. These materials attract significant attention for their ability to function as optical probes and transducers, enabling precise and sensitive detection in complex biological environments. This review highlights recent advancements in developing pDA-based optical probes, emphasizing strategies for fine-tuning synthetic parameters to optimize material properties, clarifying the fundamental sensing mechanisms underlying pDA-based systems, and exploring their potential roles in addressing global healthcare challenges. By facilitating early disease detection, real-time monitoring, and targeted therapeutic intervention, pDA-based optical probes offer transformative solutions to pressing biomedical needs. Through a comprehensive examination of cutting-edge research, this review aims to illuminate how the unique attributes of pDA materials drive innovation in biosensing technologies and contribute to improved healthcare outcomes.
Collapse
Affiliation(s)
- Jena Subhra Sulipta
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| | - Haejin Jeong
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| | - Seonki Hong
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| |
Collapse
|
3
|
Peng J, Li B, Ma Z, Qiu Z, Hu H, Jiang Y, Gao D. A microfluidic-based chemiluminescence biosensor for sensitive multiplex detection of exosomal microRNAs based on hybridization chain reaction. Talanta 2025; 281:126838. [PMID: 39255623 DOI: 10.1016/j.talanta.2024.126838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
The analysis of microRNAs (miRNAs) in exosomes is of great importance for noninvasive early disease diagnosis. However, current techniques to detect exosomal miRNAs is hampered either by laborious exosome isolation or low abundance of miRNAs in exosomes. Here, we developed a microfluidic chemiluminescence (CL) analysis method for the multiplexed detection of exosomal miR-21 and miR-155. The microfluidic device contained three parts: a snake-shaped channel for fully mixing chemiluminescent reagents, a ship-shaped channel modified with CD63 protein aptamer for capturing exosomes, and another two parallel ship-shaped channels for hybridization chain reaction (HCR) amplification and CL detection. The multiple signal amplification was realized by Y-shaped arrays, HCR amplification, and poly-HRP catalyzed CL reaction. Using this multiple signal amplification method, our microfluidic CL biosensor achieves a limit of detection of miRNAs of 0.49 fM, with a linear range of 1 fM-10 pM, which is better or comparable to previously reported biosensors. What's more, the proposed microfluidic biosensor exhibits great specificity and selectivity to the target miRNA. Moreover, the microfluidic CL strategy exhibited excellent accuracy and could significantly distinguish different cancer subtypes as well as cancer patients and healthy people. These results suggest that this simple, high sensitive, and more accurate analytical strategy by analyzing different types of exosomal miRNAs has the potential applications in cancer diagnosis and stage monitoring.
Collapse
Affiliation(s)
- Jie Peng
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Key Laboratory of Metabolomics at Shenzhen, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Baicheng Li
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Key Laboratory of Metabolomics at Shenzhen, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Zhiyuan Ma
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Key Laboratory of Metabolomics at Shenzhen, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Zhengxuan Qiu
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Key Laboratory of Metabolomics at Shenzhen, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China
| | - Hong Hu
- Division of Breast Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Yuyang Jiang
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Dan Gao
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China; Key Laboratory of Metabolomics at Shenzhen, Graduate School at Shenzhen, Tsinghua University, Shenzhen, 518055, China.
| |
Collapse
|
4
|
Syed ZU, Samaraweera S, Wang Z, Schrader JK, Scott C, Schut J, Smith DJ, Ramsey JD, Krishnan S. Colorimetric Hybridization Sensor for DNA Mimic of a SARS-CoV-2 RNA Marker: Direct and Inverse Bioanalysis. ACS MEASUREMENT SCIENCE AU 2024; 4:689-694. [PMID: 39713035 PMCID: PMC11659999 DOI: 10.1021/acsmeasuresciau.4c00043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 12/24/2024]
Abstract
This article presents a colorimetric visual biosensor designed for direct application in undiluted biofluids, which holds significant promise for point-of-need applications. Unlike traditional biosensors that struggle with heavily diluted sample matrices, the presented biosensor does not require any instrumentation or trained personnel, making it highly practical. The sensor features an oligonucleotide probe covalently attached to magnetically separable magnetite (Fe3O4) particles. This probe selectively captures a DNA mimic of the SARS-CoV-2 RNA sequence via a base-pair hybridization. The DNA mimic oligomer sequence was tested in a buffer solution, undiluted serum, and undiluted salivary biofluids. A second complementary hybridization sequence with a biotin tag was used to bind the target oligomer already hybridized to the magnetic particle-conjugated capture probe. Subsequent detection of the target oligomer was accomplished through high-affinity selective binding of streptavidin-peroxidase labels with the detection probe biotin units for visual colorimetric detection in the presence of 3,3',5,5'-tetramethylbenzidine and hydrogen peroxide. Inverse assaying of the unbound-free streptavidin-peroxidase labels left in the detection reagent solution offered a reverse trend to the target oligomer concentration, as anticipated. We obtained detection limits of 1 fM (buffer assay), 1 pM (undiluted serum assay), and 1 pM (undiluted saliva assay) and with the linear ranges of 1 fM-10 nM (buffer assay), 1 pM-1 nM (undiluted serum assay), and 1 pM-1 nM (undiluted saliva assay), respectively. The assays in different biofluids allowed for the estimation of the analytical performance and the effect of sample matrices on the detection limits and calibration sensitivity.
Collapse
Affiliation(s)
- Zia ul
Quasim Syed
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Sathya Samaraweera
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Zhuo Wang
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - James Kelby Schrader
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Colton Scott
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Joshua Schut
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Dozier Johnson Smith
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Joshua D. Ramsey
- School
of Chemical Engineering, Oklahoma State
University, Stillwater, Oklahoma 74078, United States
| | - Sadagopan Krishnan
- Department
of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| |
Collapse
|
5
|
Liu X, Fang Y, Liu J, Chen X, Teng F, Li C. Nanozyme-Based Pump-free Microfluidic Chip for Colorectal Cancer Diagnosis via Circulating Cancer Stem Cell Detection. ACS Sens 2024; 9:5090-5098. [PMID: 39373689 DOI: 10.1021/acssensors.4c00774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Circulating cancer stem cells (CCSCs) are subpopulations of cancer cells with high tumorigenicity, chemoresistance, and metastatic potential, which are also major drivers of disease progression. Herein, to achieve the prediction of tumor diagnosis and progression in colorectal cancer (CRC), a new, automated, and portable lateral displacement patterned pump-free (LP) microfluidic chip (LP-chip) with the CoPt3 nanozyme was established for CCSC capture and detection in peripheral blood and feces samples ex vivo. In this design, CoPt3@HA probes with functions of magnetic separation and colorimetric signal transduction by peroxidase-mimicking activity were applied for the capture of CCSCs and signal output in clinical samples. The generated colors of polydopamine (PDA) were quantifiable through the smartphone APP and visualizable by the naked eye in the test line (T line) and control line (C line) of the LP-chip. In the optimal experimental conditions, the CCSC concentration was sensitive to change in the range 0-105 cells mL-1, with a detection limit of 3 cells mL-1 (S/N = 3). Preliminary studies of clinical samples suggest that the platform has the potential for prediction of colorectal cancer progression and poor prognosis. Overall, the LP-chip provides potential strategies for timely diagnosis, therapeutic monitoring, and recurrence prediction to improve home-based patient care.
Collapse
Affiliation(s)
- Xiaoya Liu
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, China
- Cell and Biomolecule Recognition Research Center, School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Yiwei Fang
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, China
- Cell and Biomolecule Recognition Research Center, School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaxin Liu
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, China
- Cell and Biomolecule Recognition Research Center, School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Xinhe Chen
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, China
- Cell and Biomolecule Recognition Research Center, School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Fengmeng Teng
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Caolong Li
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 211198, China
- Cell and Biomolecule Recognition Research Center, School of Science, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
6
|
Qin M, Hu J, Li X, Liu J, Jiang R, Shi Y, Wang Z, Zhang L, Zhao Y, Gao H, Zhang Q, Zhao H, Li M, Huang C. Exosomal membrane proteins analysis using a silicon nanowire field effect transistor biosensor. Talanta 2024; 278:126534. [PMID: 39002259 DOI: 10.1016/j.talanta.2024.126534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Exosomes are of great significance in clinical diagnosis, due to their high homology with parental generation, which can reflect the pathophysiological status. However, the quantitative and classification detection of exosomes is still faced with the challenges of low sensitivity and complex operation. In this study, we develop an electrical and label-free method to directly detect exosomes with high sensitivity based on a Silicon nanowire field effect transistor biosensor (Si-NW Bio-FET). First, the impact of Debye length on Si-NW Bio-FET detection was investigated through simulation. The simulation results demonstrated that as the Debye length increased, the electrical response to Si-NW produced by charged particle at a certain distance from the surface of Si-NW was greater. A Si-NW Bio-FET modified with specific antibody CD81 on the nanowire was fabricated then used for detection of cell line-derived exosomes, which achieved a low limit of detection (LOD) of 1078 particles/mL in 0.01 × PBS. Furthermore, the Si-NW Bio-FETs modified with specific antibody CD9, CD81 and CD63 respectively, were employed to distinguish exosomes derived from human promyelocytic leukemia (HL-60) cell line in three different states (control group, lipopolysaccharide (LPS) inflammation group, and LPS + Romidepsin (FK228) drug treatment group), which was consistent with nano-flow cytometry. This study provides a highly sensitive method of directly quantifying exosomes without labeling, indicating its potential as a tool for disease surveillance and medication instruction.
Collapse
Affiliation(s)
- Meiyan Qin
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jiawei Hu
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; North China University of Technology, Beijing 100144, China
| | - Xue Li
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Jinlong Liu
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Rui Jiang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yimin Shi
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zizhen Wang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Lingqian Zhang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Yang Zhao
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Hang Gao
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Qingzhu Zhang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Haiping Zhao
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Mingxiao Li
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China.
| | - Chengjun Huang
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China; University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
7
|
Ho KHW, Lai H, Zhang R, Chen H, Yin W, Yan X, Xiao S, Lam CYK, Gu Y, Yan J, Hu K, Shi J, Yang M. SERS-Based Droplet Microfluidic Platform for Sensitive and High-Throughput Detection of Cancer Exosomes. ACS Sens 2024; 9:4860-4869. [PMID: 39233482 DOI: 10.1021/acssensors.4c01357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Exosomes, nanosized extracellular vesicles containing biomolecular cargo, are increasingly recognized as promising noninvasive biomarkers for cancer diagnosis, particularly for their role in carrying tumor-specific molecular information. Traditional methods for exosome detection face challenges such as complexity, time consumption, and the need for sophisticated equipment. This study addresses these challenges by introducing a novel droplet microfluidic platform integrated with a surface-enhanced Raman spectroscopy (SERS)-based aptasensor for the rapid and sensitive detection of HER2-positive exosomes from breast cancer cells. Our approach utilized an on-chip salt-induced gold nanoparticles (GNPs) aggregation process in the presence of HER2 aptamers and HER2-positive exosomes, enhancing the hot spot-based SERS signal amplification. This platform achieved a limit of detection of 4.5 log10 particles/mL with a sample-to-result time of 5 min per sample. Moreover, this platform has been successfully applied for HER2 status testing in clinical samples to distinguish HER2-positive breast cancer patients from HER2-negative breast cancer patients. High sensitivity, specificity, and the potential for high-throughput screening of specific tumor exosomes make this SERS-based droplet system a potential liquid biopsy technology for early cancer diagnosis.
Collapse
Affiliation(s)
- Kwun Hei Willis Ho
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Huang Lai
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Ruolin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Haitian Chen
- Department of Hepatic Surgery and Liver Transplantation Center of The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou 510630, China
| | - Wen Yin
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Xijing Yan
- Department of Breast and Thyroid Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Shu Xiao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Ching Ying Katherine Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Yutian Gu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - JiaXiang Yan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Kunpeng Hu
- Department of Breast and Thyroid Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jingyu Shi
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China
- Joint Research Center of Biosensing and Precision Theranostics, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- Research Centre for Nanoscience and Nanotechnology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| |
Collapse
|
8
|
Nieszporek A, Wierzbicka M, Labedz N, Zajac W, Cybinska J, Gazinska P. Role of Exosomes in Salivary Gland Tumors and Technological Advances in Their Assessment. Cancers (Basel) 2024; 16:3298. [PMID: 39409917 PMCID: PMC11475412 DOI: 10.3390/cancers16193298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Backgroud: Salivary gland tumors (SGTs) are rare and diverse neoplasms, presenting significant challenges in diagnosis and management due to their rarity and complexity. Exosomes, lipid bilayer vesicles secreted by almost all cell types and present in all body fluids, have emerged as crucial intercellular communication agents. They play multifaceted roles in tumor biology, including modulating the tumor microenvironment, promoting metastasis, and influencing immune responses. Results: This review focuses on the role of exosomes in SGT, hypothesizing that novel diagnostic and therapeutic approaches can be developed by exploring the mechanisms through which exosomes influence tumor occurrence and progression. By understanding these mechanisms, we can leverage exosomes as diagnostic and prognostic biomarkers, and target them for therapeutic interventions. The exploration of exosome-mediated pathways contributing to tumor progression and metastasis could lead to more effective treatments, transforming the management of SGT and improving patient outcomes. Ongoing research aims to elucidate the specific cargo and signaling pathways involved in exosome-mediated tumorigenesis and to develop standardized techniques for exosome-based liquid biopsies in clinical settings. Conclusions: Exosome-based liquid biopsies have shown promise as non-invasive, real-time systemic profiling tools for tumor diagnostics and prognosis, offering significant potential for enhancing patient care through precision and personalized medicine. Methods like fluorescence, electrochemical, colorimetric, and surface plasmon resonance (SPR) biosensors, combined with artificial intelligence, improve exosome analysis, providing rapid, precise, and clinically valid cancer diagnostics for difficult-to-diagnose cancers.
Collapse
Affiliation(s)
- Artur Nieszporek
- Biobank Research Group, Łukasiewicz Research Network–PORT Polish Centre for Technology Development, Stablowicka Street 147, 54-066 Wroclaw, Poland
| | - Małgorzata Wierzbicka
- Institute of Human Genetics Polish Academy of Sciences, Strzeszynska 32, 60-479 Poznan, Poland
- Department of Otolaryngology, Regional Specialist Hospital Wroclaw, Research & Development Centre, Kamienskiego Street 73a, 51-124 Wroclaw, Poland
| | - Natalia Labedz
- Biobank Research Group, Łukasiewicz Research Network–PORT Polish Centre for Technology Development, Stablowicka Street 147, 54-066 Wroclaw, Poland
| | - Weronika Zajac
- Faulty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland
- Materials Science and Engineering Center, Łukasiewicz Research Network–PORT Polish Centre for Technology Development, Stablowicka Street 147, 54-066 Wroclaw, Poland
| | - Joanna Cybinska
- Faulty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland
- Materials Science and Engineering Center, Łukasiewicz Research Network–PORT Polish Centre for Technology Development, Stablowicka Street 147, 54-066 Wroclaw, Poland
| | - Patrycja Gazinska
- Biobank Research Group, Łukasiewicz Research Network–PORT Polish Centre for Technology Development, Stablowicka Street 147, 54-066 Wroclaw, Poland
| |
Collapse
|
9
|
Küçük B, Yilmaz EG, Aslan Y, Erdem Ö, Inci F. Shedding Light on Cellular Secrets: A Review of Advanced Optical Biosensing Techniques for Detecting Extracellular Vesicles with a Special Focus on Cancer Diagnosis. ACS APPLIED BIO MATERIALS 2024; 7:5841-5860. [PMID: 39175406 PMCID: PMC11409220 DOI: 10.1021/acsabm.4c00782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
In the relentless pursuit of innovative diagnostic tools for cancer, this review illuminates the cutting-edge realm of extracellular vesicles (EVs) and their biomolecular cargo detection through advanced optical biosensing techniques with a primary emphasis on their significance in cancer diagnosis. From the sophisticated domain of nanomaterials to the precision of surface plasmon resonance, we herein examine the diverse universe of optical biosensors, emphasizing their specified applications in cancer diagnosis. Exploring and understanding the details of EVs, we present innovative applications of enhancing and blending signals, going beyond the limits to sharpen our ability to sense and distinguish with greater sensitivity and specificity. Our special focus on cancer diagnosis underscores the transformative potential of optical biosensors in early detection and personalized medicine. This review aims to help guide researchers, clinicians, and enthusiasts into the captivating domain where light meets cellular secrets, creating innovative opportunities in cancer diagnostics.
Collapse
Affiliation(s)
- Beyza
Nur Küçük
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Eylul Gulsen Yilmaz
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Yusuf Aslan
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Özgecan Erdem
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
| | - Fatih Inci
- UNAM—National
Nanotechnology Research Center, Bilkent
University, 06800 Ankara, Turkey
- Institute
of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| |
Collapse
|
10
|
Xu X, Zhang Z, Shen T, Pan H, Chang D. Visual dual-mode aptasensor for non-small cell lung cancer exosome detection via HRP self-coupling enhanced oxidized iridium nanoparticle aggregation. ANAL SCI 2024; 40:1459-1473. [PMID: 38900232 DOI: 10.1007/s44211-024-00582-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/15/2024] [Indexed: 06/21/2024]
Abstract
The main reason for the high mortality rate of non-small cell lung cancer is that patients are usually diagnosed at an advanced stage of the disease. Exosomes, small membrane vesicles secreted by normal cells or tumor cells, play a significant role in the progression of NSCLC. This study successfully optimized the preparation of artificial nanoenzymes self-coupling with horseradish peroxidase (IrO2NPs@HRP-AptCD63), without adding any ligand, demonstrating remarkable catalytic activity suitable for detecting the EGFR protein on the surface of NSCLC exosomes. When fused with the CD63 aptamer for identifying NSCLC exosomes, IrO2NPs@HRP showed enhanced catalytic activity in the 3,3',5,5'-tetramethylbenzidine-H2O2 oxidation-reduction system, thereby enhancing the colorimetric signal. This phenomenon can be distinguished by the naked eye and quantified using a UV-Vis spectrophotometer. Meanwhile, as the redox reaction occurs, the current signal of 3,3',5,5'-tetramethylbenzidine-H2O2, acting as an electrolyte, changes. The developed aptasensor generates dual-mode signal outputs, firstly, to visually assess the samples for their positive or negative status, and subsequently employ more in-depth electrochemical or colorimetric analysis methods for a detailed quantitative analysis of suspected positive samples. The detection limits of electrochemical analysis and colorimetric analysis were 0.9 × 103 particles/mL and 0.14 × 103 particles/mL, respectively. Compared with traditional biomarkers such as CA125, this method exhibits exceptional specificity, capable of simultaneously distinguishing serum exosomes of healthy volunteers, COPD patients, and NSCLC patients, promoting exosome detection in mouse models for tumor monitoring. Additionally, it elucidates the changes in EGFR protein expression on the surface of serum exosomes throughout the developmental trajectory.
Collapse
Affiliation(s)
- Xin Xu
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Ze Zhang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Tong Shen
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Hebei Medical University, Hebei, 050011, China
| | - Hongzhi Pan
- Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201399, China.
| | - Dong Chang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
11
|
Chowdhury R, Eslami S, Pham CV, Rai A, Lin J, Hou Y, Greening DW, Duan W. Role of aptamer technology in extracellular vesicle biology and therapeutic applications. NANOSCALE 2024; 16:11457-11479. [PMID: 38856692 DOI: 10.1039/d4nr00207e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived nanosized membrane-bound vesicles that are important intercellular signalling regulators in local cell-to-cell and distant cell-to-tissue communication. Their inherent capacity to transverse cell membranes and transfer complex bioactive cargo reflective of their cell source, as well as their ability to be modified through various engineering and modification strategies, have attracted significant therapeutic interest. Molecular bioengineering strategies are providing a new frontier for EV-based therapy, including novel mRNA vaccines, antigen cross-presentation and immunotherapy, organ delivery and repair, and cancer immune surveillance and targeted therapeutics. The revolution of EVs, their diversity as biocarriers and their potential to contribute to intercellular communication, is well understood and appreciated but is ultimately dependent on the development of methods and techniques for their isolation, characterization and enhanced targeting. As single-stranded oligonucleotides, aptamers, also known as chemical antibodies, offer significant biological, chemical, economic, and therapeutic advantages in terms of their size, selectivity, versatility, and multifunctional programming. Their integration into the field of EVs has been contributing to the development of isolation, detection, and analysis pipelines associated with bioengineering strategies for nano-meets-molecular biology, thus translating their use for therapeutic and diagnostic utility.
Collapse
Affiliation(s)
- Rocky Chowdhury
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia.
| | - Sadegh Eslami
- Molecular Proteomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | - Cuong Viet Pham
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Alin Rai
- Molecular Proteomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Cardiovascular Research, Translation and Implementation, and La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Jia Lin
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yingchu 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
| | - David W Greening
- Molecular Proteomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
- Department of Cardiovascular Research, Translation and Implementation, and La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Wei Duan
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia.
| |
Collapse
|
12
|
Yasamineh S, Nikben N, Hamed Ahmed M, Abdul Kareem R, Kadhim Al-Aridhy A, Hosseini Hooshiar M. Increasing the sensitivity and accuracy of detecting exosomes as biomarkers for cancer monitoring using optical nanobiosensors. Cancer Cell Int 2024; 24:189. [PMID: 38816782 PMCID: PMC11138050 DOI: 10.1186/s12935-024-03379-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/19/2024] [Indexed: 06/01/2024] Open
Abstract
The advancement of nanoscience and material design in recent times has facilitated the creation of point-of-care devices for cancer diagnosis and biomolecule sensing. Exosomes (EXOs) facilitate the transfer of bioactive molecules between cancer cells and diverse cells in the local and distant microenvironments, thereby contributing to cancer progression and metastasis. Specifically, EXOs derived from cancer are likely to function as biomarkers for early cancer detection due to the genetic or signaling alterations they transport as payload within the cancer cells of origin. It has been verified that EXOs circulate steadily in bodily secretions and contain a variety of information that indicates the progression of the tumor. However, acquiring molecular information and interactions regarding EXOs has presented significant technical challenges due to their nanoscale nature and high heterogeneity. Colorimetry, surface plasmon resonance (SPR), fluorescence, and Raman scattering are examples of optical techniques utilized to quantify cancer exosomal biomarkers, including lipids, proteins, RNA, and DNA. Many optically active nanoparticles (NPs), predominantly carbon-based, inorganic, organic, and composite-based nanomaterials, have been employed in biosensing technology. The exceptional physical properties exhibited by nanomaterials, including carbon NPs, noble metal NPs, and magnetic NPs, have facilitated significant progress in the development of optical nanobiosensors intended for the detection of EXOs originating from tumors. Following a summary of the biogenesis, biological functions, and biomarker value of known EXOs, this article provides an update on the detection methodologies currently under investigation. In conclusion, we propose some potential enhancements to optical biosensors utilized in detecting EXO, utilizing various NP materials such as silicon NPs, graphene oxide (GO), metal NPs, and quantum dots (QDs).
Collapse
Affiliation(s)
- Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran.
| | | | | | | | - Ameer Kadhim Al-Aridhy
- College of Health and Medical Technology, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | |
Collapse
|
13
|
Agnihotram R, Dhar R, Dhar D, Purushothaman K, Narasimhan AK, Devi A. Fusion of Exosomes and Nanotechnology: Cutting-Edge Cancer Theranostics. ACS APPLIED NANO MATERIALS 2024; 7:8489-8506. [DOI: 10.1021/acsanm.4c01033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Rohan Agnihotram
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Debolina Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Kaavya Purushothaman
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Ashwin Kumar Narasimhan
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| |
Collapse
|
14
|
Liu Q, Zheng J, Xie A, Chen M, Gong RY, Sheng Y, Chen HL, Qi CB. Exosome, a Rising Biomarkers in Liquid Biopsy: Advances of Label-Free and Label Strategy for Diagnosis of Cancer. Crit Rev Anal Chem 2024:1-12. [PMID: 38669199 DOI: 10.1080/10408347.2024.2339961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Cancer is commonly considered as one of the most severe diseases, posing a significant threat to human health and society due to various serious challenges. These challenges include difficulties in accurate diagnosis and a high propensity to form metastasis. Tissue biopsy remains the gold standard for diagnosing and subtyping cancer. However, concerns arise from its invasive nature and the potential risk of metastasis during these complex diagnostic procedures. Meanwhile, liquid biopsy has recently witnessed the rapid advancements with the emergence of three prominent detection biomarkers: circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and exosomes. Whereas, the very low abundance of CTCs combined with the instability of ctDNA intensify the challenges and decrease the accuracy of these two biomarkers for cancer diagnosis. While exosomes have gained widespread recognition as a promising biomarker in liquid biopsy due to their relatively low-invasive detection method, excellent biostability, rich resources, high abundance, and ability to provide valuable information about cancer. Therefore, it is crucial to systematically summarize recent advancements mainly in exosome-based detection methods for early cancer diagnosis. Specifically, this review will primarily focus on label-based and label-free strategies for detecting cancer using exosomes. We anticipate that this comprehensive analysis will enhance readers' understanding of the significance and value of exosomes in the fields of cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Jing Zheng
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - An Xie
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Min Chen
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Rui-Yue Gong
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yuan Sheng
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Hong-Lei Chen
- Department of Pathology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Chu-Bo Qi
- Department of Pathology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
15
|
Zhao J, Guan X, Zhang S, Sha Z, Sun S. Weak Value Amplification-Based Biochip for Highly Sensitive Detection and Identification of Breast Cancer Exosomes. BIOSENSORS 2024; 14:198. [PMID: 38667191 PMCID: PMC11048322 DOI: 10.3390/bios14040198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024]
Abstract
Exosomes constitute an emerging biomarker for cancer diagnosis because they carry multiple proteins that reflect the origins of the parent cell. The highly sensitive detection of exosomes is a crucial prerequisite for the diagnosis of cancer. In this study, we report an exosome detection system based on quantum weak value amplification (WVA). The WVA detection system consists of a reflection detection light path and a Zr-ionized biochip. Zr-ionized biochips effectively capture exosomes through the specific interaction between zirconium dioxide and the phosphate groups on the lipid bilayer of exosomes. Aptamer-modified gold nanoparticles (Au NPs) are then used to specifically recognize proteins on exosomes to enhance the detection signal. The sensitivity and resolution of the detection system are 2944.07 nm/RIU and 1.22 × 10-5 RIU, respectively. The concentration of exosomes can be directly quantified by the WVA system, ranging from 105-107 particles/mL with the detection limit of 3 × 104 particles/mL. The use of Au NPs-EpCAM for the specific enhancement of breast cancer MDA-MB-231 exosomes is demonstrated. The results indicate that the WVA detection system can be a promising candidate for the detection of exosomes as tumor markers.
Collapse
Affiliation(s)
- Jingru Zhao
- Institute of Biopharmaceutical and Healthcare Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.Z.)
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaotian Guan
- Institute of Biopharmaceutical and Healthcare Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.Z.)
| | - Sihao Zhang
- Institute of Biopharmaceutical and Healthcare Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.Z.)
| | - Zhou Sha
- Institute of Biopharmaceutical and Healthcare Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.Z.)
| | - Shuqing Sun
- Institute of Biopharmaceutical and Healthcare Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.Z.)
| |
Collapse
|
16
|
Fu M, Zhou P, Sheng W, Bai Z, Wang J, Zhu X, Hua L, Pan B, Gao F. Magnetically Controlled Photothermal, Colorimetric, and Fluorescence Trimode Assay for Gastric Cancer Exosomes Based on Acid-Induced Decomposition of CP/Mn-PBA DSNBs. Anal Chem 2024; 96:4213-4223. [PMID: 38427460 DOI: 10.1021/acs.analchem.3c05550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
The accurate quantification of cancer-derived exosomes, which are emerging as promising noninvasive biomarkers for liquid biopsies in the early diagnosis of cancer, is becoming increasingly imperative. In our work, we developed a magnetically controlled photothermal, colorimetric, and fluorescence trimode aptasensor for human gastric cancer cell (SGC-7901)-derived exosomes. This sensor relied on CP/Mn-PBA DSNBs nanocomposites, created by decorating copper peroxide (CP) nanodots on polyethyleneimine-modified manganese-containing Prussian blue analogues double-shelled nanoboxes (PEI-Mn-PBA DSNBs). Through self-assembly, we attached CD63 aptamer-labeled CP/Mn-PBA DSNBs (Apt-CP/Mn-PBA DSNBs) to complementary DNA-labeled magnetic beads (cDNA-MB). During exosome incubation, these aptamers preferentially formed complexes with exosomes, and we efficiently removed the released CP/Mn-PBA DSNBs by using magnetic separation. The CP/Mn-PBA DSNBs exhibited high photoreactivity and photothermal conversion efficiency under near-infrared (NIR) light, leading to temperature variations under 808 nm irradiation, correlating with different exosome concentrations. Additionally, colorimetric detection was achieved by monitoring the color change in a 3,3',5,5'-tetramethylbenzidine (TMB) system, facilitated by PEI modification, NIR-enhanced peroxidase-like activity of CP/Mn-PBA DSNBs and their capacity to generate Cu2+ and H2O2 under acidic conditions. Moreover, in the presence of Cu2+ and ascorbic acid (AA), DNA sequences could form dsDNA-templated copper nanoparticles (CuNPs), which emitted strong fluorescence at around 575 nm. Increasing exosome concentrations correlated with decreases in temperature, absorbance, and fluorescence intensity. This trimode biosensor demonstrated satisfactory ability in differentiating gastric cancer patients from healthy individuals using human serum samples.
Collapse
Affiliation(s)
- Mengying Fu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Peng Zhou
- Department of Orthopedics, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an 223002, China
| | - Weiwei Sheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Zetai Bai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Jin Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xu Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Lei Hua
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Bin Pan
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
17
|
Kim R, Mun B, Lim S, Park C, Kim J, Lim J, Jeong H, Son HY, Rho HW, Lim EK, Haam S. Colorimetric Detection of HER2-Overexpressing-Cancer-Derived Exosomes in Mouse Urine Using Magnetic-Polydiacetylene Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307262. [PMID: 37963850 DOI: 10.1002/smll.202307262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/31/2023] [Indexed: 11/16/2023]
Abstract
Breast cancer (BC) is a major global health problem, with ≈20-25% of patients overexpressing human epidermal growth factor receptor 2 (HER2), an aggressive marker, yet access to early detection and treatment varies across countries. A low-cost, equipment-free, and easy-to-use polydiacetylene (PDA)-based colorimetric sensor is developed for HER2-overexpressing cancer detection, designed for use in low- and middle-income countries (LMICs). PDA nanoparticles are first prepared through thin-film hydration. Subsequently, hydrophilic magnetic nanoparticles and HER2 antibodies are sequentially conjugated to them. The synthesized HER2-MPDA can be concentrated and separated by a magnetic field while inheriting the optical characteristics of PDA. The specific binding of HER2 antibody in HER2-MPDA to HER2 receptor in HER2-overexpressing exosomes causes a blue-to-red color change by altering the molecular structure of the PDA backbone. This colorimetric sensor can simultaneously separate and detect HER2-overexpressing exosomes. HER2-MPDA can detect HER2-overexpressing exosomes in the culture medium of HER2-overexpressing BC cells and in mouse urine samples from a HER2-overexpressing BC mouse model. It can selectively isolate and detect only HER2-overexpressing exosomes through magnetic separation, and its detection limit is found to be 8.5 × 108 particles mL-1. This colorimetric sensor can be used for point-of-care diagnosis of HER2-overexpressing BC in LMICs.
Collapse
Affiliation(s)
- Ryunhyung Kim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Byeonggeol Mun
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seongjae Lim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chaewon Park
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jinyoung Kim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jaewook Lim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyein Jeong
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hye Young Son
- Department of Radiology, College of Medicine, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Wook Rho
- Department of Radiology, College of Medicine, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun-Kyung Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
18
|
Jiang B, Zhang T, Liu S, Sheng Y, Hu J. Polydopamine-assisted aptamer-carrying tetrahedral DNA microelectrode sensor for ultrasensitive electrochemical detection of exosomes. J Nanobiotechnology 2024; 22:55. [PMID: 38331774 PMCID: PMC10854160 DOI: 10.1186/s12951-024-02318-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Exosomes are nanoscale extracellular vesicles (30-160 nm) with endosome origin secreted by almost all types of cells, which are considered to be messengers of intercellular communication. Cancerous exosomes serve as a rich source of biomarkers for monitoring changes in cancer-related physiological status, because they carry a large number of biological macromolecules derived from parental tumors. The ultrasensitive quantification of trace amounts of cancerous exosomes is highly valuable for non-invasive early cancer diagnosis, yet it remains challenging. Herein, we developed an aptamer-carrying tetrahedral DNA (Apt-TDNA) microelectrode sensor, assisted by a polydopamine (PDA) coating with semiconducting properties, for the ultrasensitive electrochemical detection of cancer-derived exosomes. RESULTS The stable rigid structure and orientation of Apt-TDNA ensured efficient capture of suspended exosomes. Without PDA coating signal amplification strategy, the sensor has a linear working range of 102-107 particles mL-1, with LOD of ~ 69 exosomes and ~ 42 exosomes for EIS and DPV, respectively. With PDA coating, the electrochemical signal of the microelectrode is further amplified, achieving single particle level sensitivity (~ 14 exosomes by EIS and ~ 6 exosomes by DPV). CONCLUSIONS The proposed PDA-assisted Apt-TDNA microelectrode sensor, which integrates efficient exosome capture, sensitive electrochemical signal feedback with PDA coating signal amplification, provides a new avenue for the development of simple and sensitive electrochemical sensing techniques in non-invasive cancer diagnosis and monitoring treatment response.
Collapse
Affiliation(s)
- Bowen Jiang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Tenghua Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Silan Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Yan Sheng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| | - Jiaming Hu
- International Joint Laboratory of Catalytic Chemistry, State Key Laboratory of Advanced Special Steel, Innovation Institute of Carbon Neutrality, College of Sciences, Shanghai University, Shanghai, 200444, China.
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
19
|
Ma X, Chen Z, Chen W, Chen Z, Meng X. Exosome subpopulations: The isolation and the functions in diseases. Gene 2024; 893:147905. [PMID: 37844851 DOI: 10.1016/j.gene.2023.147905] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/26/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Exosomes are nanoscale extracellular vesicles secreted by cells. Exosomes mediate intercellular communication by releasing their bioactive contents (e.g., DNAs, RNAs, lipids, proteins, and metabolites). The components of exosomes are regulated by the producing cells of exosomes. Due to their diverse origins, exosomes are highly heterogeneous in size, content, and function. Depending on these characteristics, exosomes can be divided into multiple subpopulations which have different functions. Efficient enrichment of specific subpopulations of exosomes helps to investigate their biological functions. Accordingly, numerous techniques have been developed to isolate specific subpopulations of exosomes. This review systematically introduces emerging new technologies for the isolation of different exosome subpopulations and summarizes the critical role of specific exosome subpopulations in diseases, especially in tumor occurrence and progression.
Collapse
Affiliation(s)
- Xinyi Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, China
| | - Zhenhua Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, China
| | - Wei Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, China
| | - Ziyuan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, China
| | - Xiaodan Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Thoracic Surgery, The First Affiliated Hospital of Ningbo University, Ningbo University, China.
| |
Collapse
|
20
|
Sun Z, Zhang B, Tu H, Pan C, Chai Y, Chen W. Advances in colorimetric biosensors of exosomes: novel approaches based on natural enzymes and nanozymes. NANOSCALE 2024; 16:1005-1024. [PMID: 38117141 DOI: 10.1039/d3nr05459d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Exosomes are 30-150 nm vesicles derived from diverse cell types, serving as one of the most important biomarkers for early diagnosis and prognosis of diseases. However, the conventional detection method for exosomes faces significant challenges, such as unsatisfactory sensitivity, complicated operation, and the requirement of complicated devices. In recent years, colorimetric exosome biosensors with a visual readout underwent rapid development due to the advances in natural enzyme-based assays and the integration of various types of nanozymes. These synthetic nanomaterials show unique physiochemical properties and catalytic abilities, enabling the construction of exosome colorimetric biosensors with novel principles. This review will illustrate the reaction mechanisms and properties of natural enzymes and nanozymes, followed by a detailed introduction of the recent advances in both types of enzyme-based colorimetric biosensors. A comparison between natural enzymes and nanozymes is made to provide insights into the research that improves the sensitivity and convenience of assays. Finally, the advantages, challenges, and future directions of enzymes as well as exosome colorimetric biosensors are highlighted, aiming at improving the overall performance from different approaches.
Collapse
Affiliation(s)
- Zhonghao Sun
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Binmao Zhang
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Hangjia Tu
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Chuye Pan
- College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518055, China.
| | - Yujuan Chai
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| | - Wenwen Chen
- Department of Biomedical Engineering, Shenzhen University Medicine School, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
21
|
Wang Y, Gao W, Sun M, Feng B, Shen H, Zhu J, Chen X, Yu S. A filter-electrochemical microfluidic chip for multiple surface protein analysis of exosomes to detect and classify breast cancer. Biosens Bioelectron 2023; 239:115590. [PMID: 37607449 DOI: 10.1016/j.bios.2023.115590] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/21/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023]
Abstract
Breast cancer (BC) is a complex disease with high variability and no specific tumor markers available for diagnosis. Exosomes contain rich maternal tumor information and are a novel non-invasive biomarker with the potential for cancer diagnosis and prognosis. However, analysis of exosomal protein markers in blood samples is challenging due to lengthy sample workups and insufficient sensitivity. To address this difficulty, we developed a novel filter-electrochemical microfluidic chip (FEMC) to detect and classify BC directly in whole blood without requiring heavy purification methods. In our system, exosome enrichment was performed using a dual filtration system. The target was directed through a curved channel onto four screen-printed electrodes (SPEs), where it was captured by the previously modified antibodies. Simultaneously, Zr-MOFs encapsulated with a large number of methylene blue molecules (MB@UiO-66) were absorbed on the surface of exosomes due to the high affinity for phosphate groups. This process leads to the amplification of electrical signals. The approach demonstrated that the utilization of BC exosome-associated tumor biomarkers (i.e., PMSA, EGFR, CD81, and CEA), enabled the classification of various BC mouse models samples and clinical BC samples. The entire FEMC assay was completed in 1 h with a limit of detection of 1 × 104 particles/mL. Thus, the FEMC assay can provide real-time detection information, allowing timely and better-informed opportunities for clinical BC diagnosis and typing.
Collapse
Affiliation(s)
- Yanlin Wang
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Zhejiang, 315211, China; Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Wenjing Gao
- Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Min Sun
- Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Bin Feng
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Zhejiang, 315211, China; Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Hao Shen
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Zhejiang, 315211, China; Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jianhua Zhu
- Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Xueqin Chen
- Department of Traditional Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Shaoning Yu
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Zhejiang, 315211, China; Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
22
|
P U A, Raj G, John J, Mohan K M, John F, George J. Aptamers: Features, Synthesis and Applications. Chem Biodivers 2023; 20:e202301008. [PMID: 37709723 DOI: 10.1002/cbdv.202301008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/16/2023]
Abstract
Aptamers have become a topic of interest among the researchers and scientists since they not only possess all of the benefits of antibodies but also possess special qualities including heat stability, low cost, and limitless uses⋅ Here we give a review about the features, applications, and challenges of aptamers and also how they are beneficial over the antibodies for biomedical applications. Their unique features make aptamers a prominent tool in therapeutics, diagnostics, biosensors and targeted drug delivery. In conclusion, aptamers represent exciting materials for a variety of applications and can be modified to improve their properties and to extend their applications in biomedical field.
Collapse
Affiliation(s)
- Aiswarya P U
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Gopika Raj
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Jinju John
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Malavika Mohan K
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Franklin John
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Jinu George
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| |
Collapse
|
23
|
Wang M, Shu J, Wang S, Lyu A, Wang Y, Huang D, Cui H. N-(4-Aminobutyl)- N-ethylisopropanol and Co 2+ Dual-Functionalized Core-Shell Fe 3O 4@Au/Ag Magnetic Nanomaterials with Strong and Stable Chemiluminescence for a Label-Free Exosome Immunosensor. Anal Chem 2023; 95:12982-12991. [PMID: 37587428 DOI: 10.1021/acs.analchem.3c03135] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Recently, magnetic beads (MBs) are moving toward chemiluminescence (CL) functional magnetic nanomaterials with a great potential for constructing label-free immunosensors. However, most of the CL-functionalized MBs suffer from scarce binding sites, easy aggregation, and leakage of CL reagents, which will ultimately affect the analytical performance of immunosensors. Herein, by using core-shell Fe3O4@Au/Ag magnetic nanomaterials as a nanoplatform, a novel N-(4-aminobutyl)-N-ethylisopropanol (ABEI) and Co2+ dual-functionalized magnetic nanomaterial, namely, Fe3O4@Au/Ag/ABEI/Co2+, with strong and stable CL emission was successfully synthesized. Its CL intensity was 36 and 3.5 times higher than that of MB@ABEI-Au/Co2+ and ABEI and Co2+ dual-functionalized chemiluminescent MBs previously reported by our group, respectively. It was found that the excellent CL performance of Fe3O4@Au/Ag/ABEI/Co2+ could be attributed to the enrichment effect of the Au/Ag shell and the synergistic enhance effect of the Au/Ag shell and Co2+. A related CL mechanism has been proposed. Afterward, based on the intense and stable CL emission of Fe3O4@Au/Ag/ABEI/Co2+, a sensitive and effective label-free CL immunosensor for exosome detection was established. It exhibited excellent analytical performance with a wide detection range of 3.1 × 103 to 3.1 × 108 particles/mL and a low detection limit of 2.1 × 103 particles/mL, which were better than the vast majority of the reported CL immunosensors. Moreover, the proposed label-free CL immunosensor was successfully used to detect exosomes in human serum samples and enabled us to distinguish healthy persons and lung cancer patients. It has the potential to be a powerful tool for exosome study and early cancer diagnosis.
Collapse
Affiliation(s)
- Manli Wang
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jiangnan Shu
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Shanshan Wang
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Aihua Lyu
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yisha Wang
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Dabing Huang
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of the University of Science and Technology of China, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Hua Cui
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
24
|
Wang Z, Zhang Z, Pan H, Chang D. An ultrasensitive aptasensor for exosomes detection based on biotin-streptavidin and MXenes. Anal Biochem 2023; 676:115233. [PMID: 37433362 DOI: 10.1016/j.ab.2023.115233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/10/2023] [Accepted: 07/03/2023] [Indexed: 07/13/2023]
Abstract
Exosomes derived from lung cancer typically contain the genetic information of the donor cells. Therefore, exosomes contribute to early diagnosis, treatment effectiveness evaluation, and prognosis assessment of cancer. Based on the biotin-streptavidin system and MXenes nanomaterial, a dual-effect amplification method had been developed to construct an ultrasensitive colorimetric aptasensor for detecting exosomes. MXenes can enhance the loading of aptamer and biotin as the high specific surface area. Biotin-streptavidin system can increase the amount of horseradish peroxidase-linked (HRP-linked) streptavidin, considerably boosting the color signal of the aptasensor. The proposed colorimetric aptasensor exhibited excellent sensitivity, with a detection limit of 42 particles μL-1 and a linear range of 102 to 107 particles μL-1. The constructed aptasensor showed satisfactory reproducibility, stability, and selectivity, confirming the promising application of exosomes in clinical cancer detection.
Collapse
Affiliation(s)
- Zhangmin Wang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China; Shanghai Center for Clinical Laboratory, Shanghai, 200126, China
| | - Ze Zhang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Hongzhi Pan
- Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201399, China.
| | - Dong Chang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
25
|
Mahmoodpour M, Kiasari BA, Karimi M, Abroshan A, Shamshirian D, Hosseinalizadeh H, Delavari A, Mirzei H. Paper-based biosensors as point-of-care diagnostic devices for the detection of cancers: a review of innovative techniques and clinical applications. Front Oncol 2023; 13:1131435. [PMID: 37456253 PMCID: PMC10348714 DOI: 10.3389/fonc.2023.1131435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
The development and rapid progression of cancer are major social problems. Medical diagnostic techniques and smooth clinical care of cancer are new necessities that must be supported by innovative diagnostic methods and technologies. Current molecular diagnostic tools based on the detection of blood protein markers are the most common tools for cancer diagnosis. Biosensors have already proven to be a cost-effective and accessible diagnostic tool that can be used where conventional laboratory methods are not readily available. Paper-based biosensors offer a new look at the world of analytical techniques by overcoming limitations through the creation of a simple device with significant advantages such as adaptability, biocompatibility, biodegradability, ease of use, large surface-to-volume ratio, and cost-effectiveness. In this review, we covered the characteristics of exosomes and their role in tumor growth and clinical diagnosis, followed by a discussion of various paper-based biosensors for exosome detection, such as dipsticks, lateral flow assays (LFA), and microfluidic paper-based devices (µPADs). We also discussed the various clinical studies on paper-based biosensors for exosome detection.
Collapse
Affiliation(s)
- Mehrdad Mahmoodpour
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Bahman Abedi Kiasari
- Virology Department, Faculty of Veterinary, The University of Tehran, Tehran, Iran
| | - Merat Karimi
- Institute of Nanoscience and Nanotechnology, University of Kashan, Kashan, Iran
| | - Arezou Abroshan
- Student Research Committee, Faculty of Veterinary Medicine, Shahid Bahonar University, Kerman, Iran
| | - Danial Shamshirian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Alireza Delavari
- Student's Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
26
|
Ding Z, Wei Y, Han F, Zhang X, Xu Z. DNA-Driven Photothermal Amplification Transducer for Highly Sensitive Visual Determination of Extracellular Vesicles. ACS Sens 2023; 8:2282-2289. [PMID: 37246908 DOI: 10.1021/acssensors.3c00247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Extracellular vesicles (EVs) are crucial focus of current biomedical research and future medical diagnosis. However, the requirement for specialized sophisticated instruments for quantitative readouts has limited the sensitive measurement of EVs to specialized laboratory settings, which in turn has limited bench-to-bedside translation of EV-based liquid biopsies. In this work, a straightforward temperature-output platform based on a DNA-driven photothermal amplification transducer was developed for the highly sensitive visual detection of EVs using a simple household thermometer. The EVs were specifically recognized by the antibody-aptamer sandwich immune-configuration that was constructed on portable microplates. Via a one-pot reaction, cutting-mediated exponential rolling circle amplification was initiated in situ on the EV surface, generating substantial G-quadruplex-DNA-hemin conjugates. Significant amplification in temperature was achieved from the effective photothermal conversion and regulation guided by the G-quadruplex-DNA-hemin conjugates in the 3,3',5,5'-tetramethylbenzidine-H2O2 system. Through obvious temperature outputs, the DNA-driven photothermal transducer enabled highly sensitive EV detection at close to the single-particle level and supported the highly specific identification of tumor-derived EVs directly in serum samples, without the requirement of any sophisticated instrument or labeling process. Benefiting from highly sensitive visual quantification, an easy-to-use readout, and portable detection, this photothermometric strategy is expected to be deliverable across professional on-site screening to home self-testing as EV-based liquid biopsies.
Collapse
Affiliation(s)
- Ziling Ding
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, China
| | - Yunyun Wei
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, China
| | - Fei Han
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, China
| | - Xu Zhang
- Department of Oncology and Hematology, The Third Affiliated Hospital of Shenyang Medical College, Shenyang 110034, China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang 110819, China
| |
Collapse
|
27
|
Afridi W, Strachan S, Kasetsirikul S, Pannu AS, Soda N, Gough D, Nguyen NT, Shiddiky MJA. Potential Avenues for Exosomal Isolation and Detection Methods to Enhance Small-Cell Lung Cancer Analysis. ACS MEASUREMENT SCIENCE AU 2023; 3:143-161. [PMID: 37360040 PMCID: PMC10288614 DOI: 10.1021/acsmeasuresciau.2c00068] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 06/28/2023]
Abstract
Around the world, lung cancer has long been the main factor in cancer-related deaths, with small-cell lung cancer (SCLC) being the deadliest form of lung cancer. Cancer cell-derived exosomes and exosomal miRNAs are considered promising biomarkers for diagnosing and prognosis of various diseases, including SCLC. Due to the rapidity of SCLC metastasis, early detection and diagnosis can offer better diagnosis and prognosis and therefore increase the patient's chances of survival. Over the past several years, many methodologies have been developed for analyzing non-SCLC-derived exosomes. However, minimal advances have been made in SCLC-derived exosome analysis methodologies. This Review discusses the epidemiology and prominent biomarkers of SCLC. Followed by a discussion about the effective strategies for isolating and detecting SCLC-derived exosomes and exosomal miRNA, highlighting the critical challenges and limitations of current methodologies. Finally, an overview is provided detailing future perspectives for exosome-based SCLC research.
Collapse
Affiliation(s)
- Waqar
Ahmed Afridi
- School
of Environment and Science, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Simon Strachan
- School
of Environment and Science, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Surasak Kasetsirikul
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Amandeep Singh Pannu
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Narshone Soda
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Daniel Gough
- Centre
for Cancer Research, Hudson Institute of
Medical Research, Clayton, Vic 3168, Australia
- Department
of Molecular and Translational Science, Monash University, Clayton, Vic 3168, Australia
| | - Nam-Trung Nguyen
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Muhammad J. A. Shiddiky
- School
of Environment and Science, Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Nathan Campus, Nathan, QLD 4111, Australia
| |
Collapse
|
28
|
Kumar RR, Kumar A, Chuang CH, Shaikh MO. Recent Advances and Emerging Trends in Cancer Biomarker Detection Technologies. Ind Eng Chem Res 2023. [DOI: 10.1021/acs.iecr.2c04097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Rajkumar Rakesh Kumar
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Amit Kumar
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Cheng-Hsin Chuang
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Muhammad Omar Shaikh
- Sustainability Science and Management, Tunghai University, Taichung 407224, Taiwan
| |
Collapse
|
29
|
Shaabani N, Meira SR, Marcet-Palacios M, Kulka M. Multiparametric Biosensors for Characterizing Extracellular Vesicle Subpopulations. ACS Pharmacol Transl Sci 2023; 6:387-398. [PMID: 36926451 PMCID: PMC10012251 DOI: 10.1021/acsptsci.2c00207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Indexed: 02/09/2023]
Abstract
Extracellular vesicles (EVs) are an important intercellular communication conduit for cells that have applications in precision therapy and targeted drug delivery. Small EVs, or exosomes, are a 30-150 nm phospholipid-encased subpopulation of EVs that are particularly difficult to characterize due to their small size and because they are difficult to isolate using conventional methods. In this review, we discuss some recent advances in exosome isolation, purification, and sensing platforms using microfluidics, acoustics, and size exclusion chromatography. We discuss some of the challenges and unanswered questions with respect to understanding exosome size heterogeneity and how modern biosensor technology can be applied to exosome isolation. In addition, we discuss how some advancements in sensing platforms such as colorimetric, fluorescent, electronic, surface plasmon resonance (SPR), and Raman spectroscopy may be applied to exosome detection in multiparametric systems. The application of cryogenic electron tomography and microscopy to understanding exosome ultrastructure will become vital as this field progresses. In conclusion, we speculate on some future needs in the exosome research field and how these technologies could be applied.
Collapse
Affiliation(s)
- Narges Shaabani
- Nanotechnology
Research Centre, National Research Council
Canada, Edmonton, Alberta T6G 2M9, Canada
| | - Sabrina Rodrigues Meira
- Nanotechnology
Research Centre, National Research Council
Canada, Edmonton, Alberta T6G 2M9, Canada
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | | | - Marianna Kulka
- Nanotechnology
Research Centre, National Research Council
Canada, Edmonton, Alberta T6G 2M9, Canada
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
30
|
Del Real Mata C, Jeanne O, Jalali M, Lu Y, Mahshid S. Nanostructured-Based Optical Readouts Interfaced with Machine Learning for Identification of Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2202123. [PMID: 36443009 DOI: 10.1002/adhm.202202123] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/14/2022] [Indexed: 11/30/2022]
Abstract
Extracellular vesicles (EVs) are shed from cancer cells into body fluids, enclosing molecular information about the underlying disease with the potential for being the target cancer biomarker in emerging diagnosis approaches such as liquid biopsy. Still, the study of EVs presents major challenges due to their heterogeneity, complexity, and scarcity. Recently, liquid biopsy platforms have allowed the study of tumor-derived materials, holding great promise for early-stage diagnosis and monitoring of cancer when interfaced with novel adaptations of optical readouts and advanced machine learning analysis. Here, recent advances in labeled and label-free optical techniques such as fluorescence, plasmonic, and chromogenic-based systems interfaced with nanostructured sensors like nanoparticles, nanoholes, and nanowires, and diverse machine learning analyses are reviewed. The adaptability of the different optical methods discussed is compared and insights are provided into prospective avenues for the translation of the technological approaches for cancer diagnosis. It is discussed that the inherent augmented properties of nanostructures enhance the sensitivity of the detection of EVs. It is concluded by reviewing recent integrations of nanostructured-based optical readouts with diverse machine learning models as novel analysis ventures that can potentially increase the capability of the methods to the point of translation into diagnostic applications.
Collapse
Affiliation(s)
| | - Olivia Jeanne
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Mahsa Jalali
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Yao Lu
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Sara Mahshid
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| |
Collapse
|
31
|
Woldekidan HB, Woldesemayat AA, Adam G, Tafesse M, Thimiri Govinda Raj DB. Aptamer-Based Tumor-Targeted Diagnosis and Drug Delivery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:173-192. [PMID: 35896892 DOI: 10.1007/5584_2022_732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Early cancer identification is crucial for providing patients with safe and timely therapy. Highly dependable and adaptive technologies will be required to detect the presence of biological markers for cancer at very low levels in the early stages of tumor formation. These techniques have been shown to be beneficial in encouraging patients to develop early intervention plans, which could lead to an increase in the overall survival rate of cancer patients. Targeted drug delivery (TDD) using aptamer is promising due to its favorable properties. Aptamer is suitable for superior TDD system candidates due to its desirable properties including a high binding affinity and specificity, a low immunogenicity, and a chemical composition that can be simply changed.Due to these properties, aptamer-based TDD application has limited drug side effect along with organ damages. The development of aptasensor has been promising in TDD for cancer cell treatment. There are biomarkers and expressed molecules during cancer cell development; however, only few are addressed in aptamer detection study of those molecules. Its great potential of attachment of binding to specific target molecule made aptamer a reliable recognition element. Because of their unique physical, chemical, and biological features, aptamers have a lot of potential in cancer precision medicine.In this review, we summarized aptamer technology and its application in cancer. This includes advantages properties of aptamer technology over other molecules were thoroughly discussed. In addition, we have also elaborated the application of aptamer as a direct therapeutic function and as a targeted drug delivery molecule (aptasensor) in cancer cells with several examples in preclinical and clinical trials.
Collapse
Affiliation(s)
- Haregewoin Bezu Woldekidan
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Biotechnology, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Adugna A Woldesemayat
- Department of Biotechnology, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Getachew Adam
- Sustainable Energy Center of Excellence, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Mesfin Tafesse
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Council for Scientific and Industrial Research, Pretoria, South Africa
- Biotechnology and Bioprocessing Center of Excellence, Addis Ababa Science and Technology University, Addis Ababa, Ethiopia
| | - Deepak B Thimiri Govinda Raj
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Council for Scientific and Industrial Research, Pretoria, South Africa.
| |
Collapse
|
32
|
Liu J, Xu JZ, Rao ZM, Zhang WG. An enzymatic colorimetric whole-cell biosensor for high-throughput identification of lysine overproducers. Biosens Bioelectron 2022; 216:114681. [PMID: 36087402 DOI: 10.1016/j.bios.2022.114681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/17/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
L-lysine is a crucial nutrient for both humans and animals, and its main commercial use is as a supplement in animal feed to promote chicken and other animal growth. Fluorescence biosensors based on the transcriptional regulator have been developed for high-throughput screening of L-lysine producers. However, due to its inability to specifically detect lysine, this fluorescent biosensor cannot be employed to screen high-yielding strains. Here, we present a novel technique for observing L-lysine concentrations within individual Corynebacterium glutamicum cells. The transcriptional regulator LysG and its binding site, as well as the phytoene desaturase that catalyzes the synthesis of the red pigment, make up the functional core of the biosensor. The lysine-sensitive mutant LysG(E123Y, E125A), which improved the sensitivity of biosensors, was generated by site-directed saturation mutagenesis. In addition, we increased the lysine-induced chromogenic biosensor response to 320 mM by optimizing the L-lysine export mechanism and the pathway for the synthesis of lycopene precursors. The direct identification of producers with elevated L-lysine accumulation is thus made straightforward by colorimetric screening. Lys-8, a lysine producer with a maximum lysine titer of 316.2 mM, was sorted out based on the biosensor. The enzymatic colorimetric biosensor constructed here is a simple tool with great potential for the development of high-level lysine-producing C. glutamicum.
Collapse
Affiliation(s)
- Jie Liu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800(#)Lihu Road, WuXi, 214122, PR China
| | - Jian-Zhong Xu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800(#)Lihu Road, WuXi, 214122, PR China
| | - Zhi-Ming Rao
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800(#)Lihu Road, WuXi, 214122, PR China; National Engineering Laboratory for Cereal Fermentation Technology, School of Biotechnology, Jiangnan University, 1800(#)Lihu Road, WuXi, 214122, PR China
| | - Wei-Guo Zhang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800(#)Lihu Road, WuXi, 214122, PR China.
| |
Collapse
|
33
|
Tang Q, Xiao X, Li R, He H, Li S, Ma C. Recent Advances in Detection for Breast-Cancer-Derived Exosomes. Molecules 2022; 27:molecules27196673. [PMID: 36235208 PMCID: PMC9571663 DOI: 10.3390/molecules27196673] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/28/2022] Open
Abstract
Breast cancer is the most common malignant tumor in women, its incidence is secret, and more than half of the patients are diagnosed in the middle and advanced stages, so it is necessary to develop simple and efficient detection methods for breast cancer diagnosis to improve the survival rate and quality of life of breast cancer patients. Exosomes are extracellular vesicles secreted by all kinds of living cells, and play an important role in the occurrence and development of breast cancer and the formation of the tumor microenvironment. Exosomes, as biomarkers, are an important part of breast cancer fluid biopsy and have become ideal targets for the early diagnosis, curative effect evaluation, and clinical treatment of breast cancer. In this paper, several traditional exosome detection methods, including differential centrifugation and immunoaffinity capture, were summarized, focusing on the latest research progress in breast cancer exosome detection. It was summarized from the aspects of optics, electrochemistry, electrochemiluminescence and other aspects. This review is expected to provide valuable guidance for exosome detection of clinical breast cancer and the establishment of more reliable, efficient, simple and innovative methods for exosome detection of breast cancer in the future.
Collapse
Affiliation(s)
- Qin Tang
- School of Life Sciences, Central South University, Changsha 410013, China
- Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Xinying Xiao
- School of Life Sciences, Central South University, Changsha 410013, China
- Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Ranhao Li
- School of Life Sciences, Central South University, Changsha 410013, China
- Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Hailun He
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Shanni Li
- School of Life Sciences, Central South University, Changsha 410013, China
- Correspondence: (S.L.); (C.M.)
| | - Changbei Ma
- School of Life Sciences, Central South University, Changsha 410013, China
- Correspondence: (S.L.); (C.M.)
| |
Collapse
|
34
|
Lee M, Kim J, Jang M, Park C, Lee JH, Lee T. Introduction of Nanomaterials to Biosensors for Exosome Detection: Case Study for Cancer Analysis. BIOSENSORS 2022; 12:648. [PMID: 36005042 PMCID: PMC9405681 DOI: 10.3390/bios12080648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/04/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022]
Abstract
Exosomes have been gaining attention for early cancer diagnosis owing to their biological functions in cells. Several studies have reported the relevance of exosomes in various diseases, including pancreatic cancer, retroperitoneal fibrosis, obesity, neurodegenerative diseases, and atherosclerosis. Particularly, exosomes are regarded as biomarkers for cancer diagnosis and can be detected in biofluids, such as saliva, urine, peritoneal fluid, and blood. Thus, exosomes are advantageous for cancer liquid biopsies as they overcome the current limitations of cancer tissue biopsies. Several studies have reported methods for exosome isolation, and analysis for cancer diagnosis. However, further clinical trials are still required to determine accurate exosome concentration quantification methods. Recently, various biosensors have been developed to detect exosomal biomarkers, including tumor-derived exosomes, nucleic acids, and proteins. Among these, the exact quantification of tumor-derived exosomes is a serious obstacle to the clinical use of liquid biopsies. Precise detection of exosome concentration is difficult because it requires clinical sample pretreatment. To solve this problem, the use of the nanobiohybrid material-based biosensor provides improved sensitivity and selectivity. The present review will discuss recent progress in exosome biosensors consisting of nanomaterials and biomaterial hybrids for electrochemical, electrical, and optical-based biosensors.
Collapse
Affiliation(s)
- Myoungro Lee
- Department of Chemical Engineering, Kwangwoon University, Seoul 01897, Korea
| | - Jinmyeong Kim
- Department of Chemical Engineering, Kwangwoon University, Seoul 01897, Korea
| | - Moonbong Jang
- Department of Chemical Engineering, Kwangwoon University, Seoul 01897, Korea
| | - Chulhwan Park
- Department of Chemical Engineering, Kwangwoon University, Seoul 01897, Korea
| | - Jin-Ho Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Korea
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, Seoul 01897, Korea
| |
Collapse
|
35
|
Exo-III enzyme based colorimetric small extracellular vesicles (sEVs) detection via G-quadruplex-based signal quenching strategy. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
36
|
Rolling circle amplification assisted dual signal amplification colorimetric biosensor for ultrasensitive detection of leukemia-derived exosomes. Talanta 2022; 245:123444. [DOI: 10.1016/j.talanta.2022.123444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/27/2022] [Accepted: 04/02/2022] [Indexed: 11/22/2022]
|
37
|
Xu L, Ramadan S, Rosa BG, Zhang Y, Yin T, Torres E, Shaforost O, Panagiotopoulos A, Li B, Kerherve G, Kim DK, Mattevi C, Jiao LR, Petrov PK, Klein N. On-chip integrated graphene aptasensor with portable readout for fast and label-free COVID-19 detection in virus transport medium. SENSORS & DIAGNOSTICS 2022; 1:719-730. [PMID: 35923775 PMCID: PMC9280445 DOI: 10.1039/d2sd00076h] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/10/2022] [Indexed: 01/12/2023]
Abstract
Graphene field-effect transistor (GFET) biosensors exhibit high sensitivity due to a large surface-to-volume ratio and the high sensitivity of the Fermi level to the presence of charged biomolecules near the surface. For most reported GFET biosensors, bulky external reference electrodes are used which prevent their full-scale chip integration and contribute to higher costs per test. In this study, GFET arrays with on-chip integrated liquid electrodes were employed for COVID-19 detection and functionalized with either antibody or aptamer to selectively bind the spike proteins of SARS-CoV-2. In the case of the aptamer-functionalized GFET (aptasensor, Apt-GFET), the limit-of-detection (LOD) achieved was about 103 particles per mL for virus-like particles (VLPs) in clinical transport medium, outperforming the Ab-GFET biosensor counterpart. In addition, the aptasensor achieved a LOD of 160 aM for COVID-19 neutralizing antibodies in serum. The sensors were found to be highly selective, fast (sample-to-result within minutes), and stable (low device-to-device signal variation; relative standard deviations below 0.5%). A home-built portable readout electronic unit was employed for simultaneous real-time measurements of 12 GFETs per chip. Our successful demonstration of a portable GFET biosensing platform has high potential for infectious disease detection and other health-care applications.
Collapse
Affiliation(s)
- Lizhou Xu
- Department of Materials, Imperial College LondonLondonSW7 2AZUK,ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang UniversityHangzhou311200China
| | - Sami Ramadan
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| | | | - Yuanzhou Zhang
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| | - Tianyi Yin
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| | - Elias Torres
- Graphenea SemiconductorPaseo Mikeletegi 83San Sebastián20009Spain
| | - Olena Shaforost
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| | | | - Bing Li
- Department of Brain Sciences, Imperial College LondonLondonW12 0BZUK,Care Research & Technology Centre, UK Dementia Research InstituteW12 0BZUK,Institute for Materials Discovery, University College LondonRoberts BuildingLondonWC1E 7JEUK
| | | | - Dong Kuk Kim
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| | - Cecilia Mattevi
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| | - Long R. Jiao
- Department of Hepatobiliary Surgery, Division of Surgery & Cancer, Imperial College LondonHammersmith Hospital Campus, Du Cane RoadLondonW12 0NNUK
| | - Peter K. Petrov
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| | - Norbert Klein
- Department of Materials, Imperial College LondonLondonSW7 2AZUK
| |
Collapse
|
38
|
Davydova A, Vorobyeva M. Aptamer-Based Biosensors for the Colorimetric Detection of Blood Biomarkers: Paving the Way to Clinical Laboratory Testing. Biomedicines 2022; 10:biomedicines10071606. [PMID: 35884911 PMCID: PMC9313021 DOI: 10.3390/biomedicines10071606] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Clinical diagnostics for human diseases rely largely on enzyme immunoassays for the detection of blood biomarkers. Nevertheless, antibody-based test systems have a number of shortcomings that have stimulated a search for alternative diagnostic assays. Oligonucleotide aptamers are now considered as promising molecular recognizing elements for biosensors (aptasensors) due to their high affinity and specificity of target binding. At the moment, a huge variety of aptasensors have been engineered for the detection of various analytes, especially disease biomarkers. However, despite their great potential and excellent characteristics in model systems, only a few of these aptamer-based assays have been translated into practice as diagnostic kits. Here, we will review the current progress in the engineering of aptamer-based colorimetric assays as the most suitable format for clinical lab diagnostics. In particular, we will focus on aptasensors for the detection of blood biomarkers of cardiovascular, malignant, and neurodegenerative diseases along with common inflammation biomarkers. We will also analyze the main obstacles that have to be overcome before aptamer test systems can become tantamount to ELISA for clinical diagnosis purposes.
Collapse
|
39
|
Li H, Huang T, Lu L, Yuan H, Zhang L, Wang H, Yu B. Ultrasensitive Detection of Exosomes Using an Optical Microfiber Decorated with Plasmonic MoSe 2-Supported Gold Nanorod Nanointerfaces. ACS Sens 2022; 7:1926-1935. [DOI: 10.1021/acssensors.2c00598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Hongtao Li
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
- Guangxi Key Laboratory of Nuclear Physics and Nuclear Technology, College of Physics Science and Technology, Guangxi Normal University, Guilin 541004, People’s Republic of China
| | - Tianqi Huang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Liang Lu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Hao Yuan
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Lei Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| | - Hongzhi Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei 230031, People’s Republic of China
- Institute of Urology, Anhui Medical University Hefei, 230031, People’s Republic of China
| | - Benli Yu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Anhui University, Hefei 230601, People’s Republic of China
- School of Physics and Optoelectronics Engineering, Key Laboratory of Optoelectronic Information Acquisition and Manipulation of Ministry of Education, Anhui University, Hefei 230601, People’s Republic of China
| |
Collapse
|
40
|
Altuner EE, Ozalp VC, Yilmaz MD, Sudagidan M, Aygun A, Acar EE, Tasbasi BB, Sen F. Development of electrochemical aptasensors detecting phosphate ions on TMB substrate with epoxy-based mesoporous silica nanoparticles. CHEMOSPHERE 2022; 297:134077. [PMID: 35218784 DOI: 10.1016/j.chemosphere.2022.134077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/10/2022] [Accepted: 02/19/2022] [Indexed: 06/14/2023]
Abstract
This study, it is aimed to develop an electrochemical aptasensor that can detect phosphate ions using 3.3'5.5' tetramethylbenzidine (TMB). It is based on the principle of converting the binding affinity of the target molecule phosphate ion (PO43-) into an electrochemical signal with specific aptamer sequences for the aptasensor to be developed. The aptamer structure served as a gate for the TMB to be released and was used to trap the TMB molecule in mesoporous silica nanoparticles (MSNPs). The samples for this study were characterized by transmission electron spectroscopy (TEM), Brunner-Emmet-Teller, dynamic light scattering&electrophoretic light scattering, and induction coupled plasma atomic emission spectroscopy. According to TEM analysis, MSNPs have a morphologically hexagonal structure and an average size of 208 nm. In this study, palladium-carbon nanoparticles (Pd/C NPs) with catalytic reaction were used as an alternative to the biologically used horseradish peroxidase (HRP) enzyme for the release of TMB in the presence of phosphate ions. The limit of detection (LOD) was calculated as 0.983 μM, the limit of determination (LOQ) was calculated as 3.276 μM, and the dynamic linear phosphate range was found to be 50-1000 μM. The most important advantage of this bio-based aptasensor assembly is that it does not contain molecules such as a protein that cannot be stored for a long time at room temperature, so its shelf life is very long compared to similar systems developed with antibodies. The proposed sensor shows good recovery in phosphate ion detection and is considered to have great potential among electrochemical sensors.
Collapse
Affiliation(s)
- Elif Esra Altuner
- Department of Biochemistry, Dumlupinar University, 43000, Kutahya, Turkiye.
| | - Veli Cengiz Ozalp
- Medical School, Department of Medical Biology, Atilim University, 06830, Ankara, Turkiye.
| | - M Deniz Yilmaz
- Department of Bioengineering, Faculty of Engineering and Architecture, Konya Food and Agriculture University, 42080, Konya, Turkiye
| | - Mert Sudagidan
- KIT-ARGEM, R&D Center, Konya Food and Agriculture University, 42080, Konya, Turkiye
| | - Aysenur Aygun
- Department of Biochemistry, Dumlupinar University, 43000, Kutahya, Turkiye
| | - Elif Esma Acar
- KIT-ARGEM, R&D Center, Konya Food and Agriculture University, 42080, Konya, Turkiye
| | - Behiye Busra Tasbasi
- KIT-ARGEM, R&D Center, Konya Food and Agriculture University, 42080, Konya, Turkiye
| | - Fatih Sen
- Department of Biochemistry, Dumlupinar University, 43000, Kutahya, Turkiye.
| |
Collapse
|
41
|
Chen H, Bian F, Guo J, Zhao Y. Aptamer-Functionalized Barcodes in Herringbone Microfluidics for Multiple Detection of Exosomes. SMALL METHODS 2022; 6:e2200236. [PMID: 35466594 DOI: 10.1002/smtd.202200236] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/22/2022] [Indexed: 05/04/2023]
Abstract
Tumor-derived exosomes are vital for clinical dynamic and accurate tumor diagnosis, thus developing sensitive and multiple exosomes detection technology has attracted remarkable attention of scientists. Here, a novel herringbone microfluidic device with aptamer-functionalized barcodes integration for specific capture and multiple detection of tumor-derived exosomes is presented. The barcodes with core-shell constructions are obtained by partially replicating the periodically ordered hexagonal close-packaged colloidal crystal beads. As their inverse opal hydrogel shell possesses rich interconnected pores, the barcodes could provide abundant surface area for functionalization of DNA aptamers to realize specific recognition of target exosomes. Besides, the encoded structure colors of the barcodes can be maintained stably during the detection events as their hardish cores are with sufficient mechanical strength. It is demonstrated that by embedding these barcodes in herringbone groove microfluidic device with designed patterns, the specific capture efficiency and synergetic detection of multiple tumor-derived exosomes in peripheral blood can be significantly improved due to enhanced resistance of turbulent flow. These features make the aptamer-functionalized barcodes and herringbone microfluidics integrated platform promising for exosomes extraction and dynamic tumor diagnosis.
Collapse
Affiliation(s)
- Hanxu Chen
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Feika Bian
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Jiahui Guo
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| |
Collapse
|
42
|
Cheng Y, Xie Q, He M, Chen B, Chen G, Yin X, Kang Q, Xu Y, Hu B. Sensitive detection of exosomes by gold nanoparticles labeling inductively coupled plasma mass spectrometry based on cholesterol recognition and rolling circle amplification. Anal Chim Acta 2022; 1212:339938. [DOI: 10.1016/j.aca.2022.339938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 11/26/2022]
|
43
|
Khoris IM, Nasrin F, Chowdhury AD, Park EY. Advancement of dengue virus NS1 protein detection by 3D-nanoassembly complex gold nanoparticles utilizing competitive sandwich aptamer on disposable electrode. Anal Chim Acta 2022; 1207:339817. [DOI: 10.1016/j.aca.2022.339817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/18/2022] [Accepted: 04/06/2022] [Indexed: 12/25/2022]
|
44
|
Khan A, Di K, Khan H, He N, Li Z. Rapid Capturing and Chemiluminescent Sensing of Programmed Death Ligand-1 Expressing Extracellular Vesicles. BIOSENSORS 2022; 12:bios12050281. [PMID: 35624582 PMCID: PMC9138745 DOI: 10.3390/bios12050281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/17/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022]
Abstract
Cancer specific extracellular vesicles (EVs) are of significant clinical relevance, for instance programmed death ligand-1 (PD-L1) expressing EVs (PD-L1@EVs) have been shown to be ideal biomarker for non-invasive diagnosis of cancer and can predate the response of cancer patients to anti-PD-1/PD-L-1 immunotherapy. The development of sensitive and straightforward methods for detecting PD-L1@EVs can be a vital tool for non-invasive diagnosis of cancer. Most of the contemporary methods for EVs detection have limitations such as involvement of long and EV’s loss prone isolation methods prior to detection or they have employed expensive antibodies and instruments to accomplish detection. Therefore, we designed an ultracentrifugation-free and antibody-free sensing assay for PD-L1@EV by integrating Titanium oxide (TiO2) coated magnetic beads (Fe3O4@TiO2) rapid capturing of EVs from undiluted serum with aptamers specificity and chemiluminescence (CL) sensitivity. To accomplish this we used Fe3O4@TiO2 beads to rapidly capture EVs from the undiluted patient serum and added biotin labelled PD-L1 aptamer to specifically recognize PD-L1@EVs. Later, added streptavidin-modified Alkaline phosphates (ALP) taking advantage of biotin-streptavidin strong binding. Addition of CDP-star, a chemiluminescent substrate of ALP, initiates the chemiluminiscense that was recorded using spectrophotometer. The sensing assay showed high sensitivity with limit of detection (LOD) as low as 2.584×105 EVs/mL and a wider linear correlation of CL intensity (a.u.) with the concentration of PD-L1@EVs from 105 to 108 EVs/mL. To examine the clinical utility of sensing assay we used undiluted serum samples from lung cancer patients and healthy individuals and successfully discern between healthy individuals and lung cancer patients. We are optimistic that the sensing assay can ameliorate our ability to be able to diagnose lung cancer non-invasively and can be helpful to predate the patient’s response to anti-PD-1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Adeel Khan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China;
| | - Kaili Di
- Department of Clinical Laboratory Medicine, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (K.D.); (Z.L.)
| | - Haroon Khan
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China;
- Correspondence:
| | - Zhiyang Li
- Department of Clinical Laboratory Medicine, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (K.D.); (Z.L.)
| |
Collapse
|
45
|
Ding Z, Lu Y, Wei Y, Song D, Xu Z, Fang J. DNA-Engineered iron-based metal-organic framework bio-interface for rapid visual determination of exosomes. J Colloid Interface Sci 2022; 612:424-433. [PMID: 34999547 DOI: 10.1016/j.jcis.2021.12.133] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 11/28/2022]
Abstract
In this study, a rapid, low-cost and facile method for detecting exosomes was developed by engineering DNA ligands on the surface of an iron-based metal-organic framework (Fe-MOF). Aptamers of exosomal transmembrane CD63 protein (CD63-aptamers) were utilized as both the optically active layer and the exosome-specific recognition element to engineer an Fe-MOF bio-interface for high-efficiency regulation of the catalytic behavior of Fe-MOF toward the chromogenic substrate. The effective enhancement of the intrinsic peroxidase-like catalytic activity was confirmed via the self-assembly of CD63-aptamers on the surface of Fe-MOF. The specific binding of exosomes with CD63-aptamers altered the conformation of DNA ligands on the surface of Fe-MOF, contributing to sensitive variation in Fe-MOF catalytic activity. This directly produced a distinct color change and enabled the visual detection of exosomes. Via one-step "mixing-and-detection", the Fe-MOF bio-interface exhibited excellent performance in quantitative analysis of exosomes derived from human breast cancer cell lines ranging from 1.1 × 105 to 2.2 × 107 particles/μL with a detection limit of 5.2 × 104 particles/μL. The expression of exosomal CD63 proteins originated from three types of cancer cell lines, including breast cancer, gastric cancer and lung cancer cell lines, was differentiated within only 17 min. Furthermore, the method was successfully applied to the identification of exosomes in serum samples, suggesting its potential in clinical analysis as a valuable tool for the rapid, convenient and economical testing of exosomes.
Collapse
Affiliation(s)
- Ziling Ding
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China
| | - Yanbing Lu
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 110122 Shenyang, China
| | - Yunyun Wei
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China
| | - Dan Song
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, 110819 Shenyang, China.
| | - Jin Fang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 110122 Shenyang, China
| |
Collapse
|
46
|
Wei J, Zhu K, Chen Z, Yang Z, Yang K, Wang Z, Zong S, Cui Y. Triple-color fluorescence co-localization of PD-L1-overexpressing cancer exosomes. Mikrochim Acta 2022; 189:182. [PMID: 35394232 DOI: 10.1007/s00604-022-05278-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/16/2022] [Indexed: 11/28/2022]
Abstract
Programed cell death ligand 1 (PD-L1) is a protein biomarker overexpressed on exosomes derived from tumor cells. It plays an important role in tumor diagnosis, screening, evaluation of therapeutic efficacy, and prognosis. In this study, a facile method is presented to detect PD-L1-overexpressing cancer exosomes with high specificity and sensitivity. First, gold nanospheres (GNSs) were attached to the bottom of an eight-well chambered slide by electrostatic adsorption, forming the detection substrate. Then, Cy5-labeled CD63 aptamers (i.e., the capture probes) were modified on the GNSs by Au-S bond. After adding samples containing target exosomes which were stained by membrane dyes DiI in advance, FAM-labeled PD-L1 aptamers (i.e., the immunoprobes) were added to recognize PD-L1 on the target exosomes. By triple-color fluorescence co-localization (TFC) of the Cy5, DiI, and FAM channels, highly sensitive and reliable detection of the PD-L1-overexpressing exosomes was achieved in the concentration range 7.78 × 101 to 7.78 × 104 particles/mL with a detection limit down to 6 particles/mL. The advantages of the proposed detection method include the following; first, the detection substrate is easy to prepare and convenient to clean. Second, the TFC strategy can completely exclude nonspecific reaction sites and thus significantly improves the accuracy. Such a facile and reliable detection method holds a great potential in exosome-based cancer theranostics. In this paper, we proposed a triple-color fluorescence co-localization (TFC) strategy to significantly improve the reliability of exosome detection and the detection substrate is easy to prepare and convenient to clean. In addition, the LOD is down to 6 particles/mL, which is quite low compared with other detection methods.
Collapse
Affiliation(s)
- Jinxiu Wei
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Kai Zhu
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Zengwei Chen
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Zhaoyan Yang
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Kuo Yang
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Zhuyuan Wang
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China
| | - Shenfei Zong
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China.
| | - Yiping Cui
- Advanced Photonics Center, Southeast University, Nanjing, 210096, Jiangsu, China.
| |
Collapse
|
47
|
Sousa DA, Carneiro M, Ferreira D, Moreira FTC, Sales MGFV, Rodrigues LR. Recent advances in the selection of cancer-specific aptamers for the development of biosensors. Curr Med Chem 2022; 29:5850-5880. [PMID: 35209816 DOI: 10.2174/0929867329666220224155037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/30/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
An early diagnosis has the potential to greatly decrease cancer mortality. For that purpose, specific cancer biomarkers have been molecularly targeted by aptamer sequences to enable an accurate and rapid detection. Aptamer-based biosensors for cancer diagnostics are a promising alternative to those using antibodies, due to their high affinity and specificity to the target molecules and advantageous production. Synthetic nucleic acid aptamers are generated by in vitro Systematic Evolution of Ligands by Exponential enrichment (SELEX) methodologies that have been improved over the years to enhance the efficacy and to shorten the selection process. Aptamers have been successfully applied in electrochemical, optical, photoelectrochemical and piezoelectrical-based detection strategies. These aptasensors comprise a sensitive, accurate and inexpensive option for cancer detection being used as point-of-care devices. This review highlights the recent advances in cancer biomarkers, achievements and optimizations made in aptamer selection, as well as the different aptasensors developed for the detection of several cancer biomarkers.
Collapse
Affiliation(s)
- Diana A Sousa
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- MIT-Portugal Program, Lisbon, Portugal
| | - Mariana Carneiro
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- BioMark@ISEP, School of Engineering, Polytechnic of Porto, Porto, Portugal
| | - Débora Ferreira
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- MIT-Portugal Program, Lisbon, Portugal
| | - Felismina T C Moreira
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- BioMark@ISEP, School of Engineering, Polytechnic of Porto, Porto, Portugal
| | - Maria Goreti F V Sales
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
- MIT-Portugal Program, Lisbon, Portugal
- BioMark@UC, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Lígia R Rodrigues
- CEB- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| |
Collapse
|
48
|
Shi L, Cao J, Yang C, Wang X, Shi K, Shang L. Hierarchical magnetic nanoparticles for highly effective capture of small extracellular vesicles. J Colloid Interface Sci 2022; 615:408-416. [PMID: 35149353 DOI: 10.1016/j.jcis.2022.01.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/13/2022] [Accepted: 01/26/2022] [Indexed: 11/27/2022]
Abstract
Small extracellular vesicles (EVs) have various functions through the transfer of specific biomolecules. However, it is still challenging to capture small EVs with high sensitivity and specificity. Herein, inspired by the unique burry structure and the strong adhesion ability of pollen grains, we presented a novel Fe3O4@MgSiO3 hierarchical magnetic nanoparticles (HNPs) as nanocarriers for the capture of small EVs. The NPs were generated through the solvothermal method and further modified with branching dendrimers to exhibit a hierarchical morphology. The enlarged surface area facilitated high-efficient capture of small EVs through specific recognition of aptamer probes and the small EVs surface markers. Besides, the magnetic core of the NPs allowed them to be isolated under the action of an external magnetic field, and thus the captured small EVs could be easily separated from plasma. These results indicated that the HNPs could serve as excellent nanocarriers for small EVs capture and related biomedical applications.
Collapse
Affiliation(s)
- Liang Shi
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Jie Cao
- Translational Medicine Laboratory, Wound Healing and Regenerative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chaoyu Yang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Xiaocheng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Keqing Shi
- Translational Medicine Laboratory, Wound Healing and Regenerative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
49
|
Translating cancer exosomes detection into the color change of phenol red based on target-responsive DNA microcapsules. Anal Chim Acta 2022; 1192:339357. [PMID: 35057959 DOI: 10.1016/j.aca.2021.339357] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/29/2021] [Accepted: 12/05/2021] [Indexed: 11/15/2022]
Abstract
Emerging evidence indicates that exosomes can be used as a potential biomarker for monitoring diseases, including cancer. However, enhancing the sensing performance in terms of convenience and sensitivity remains an urgent demand for exosomes detection. In this study, a pH-sensitive colorimetric biosensing strategy was developed for exosomes detection by integrating stimuli-responsive DNA microcapsules and acetylcholinesterase to produce acetic acid. The constructed DNA microcapsules consisted of DNA shells crosslinked by anti-CD63 aptamers and loaded with acetylcholinesterase. With exosomes addition, an energetically stabilized aptamer-CD63 compound was produced and microcapsules dissociated due to the reaction of surface protein CD63 of exosomes and aptamer of CD63, resulting in the release of encapsulated AChE. Through a simple centrifugation separation, unreacted DNA microcapsules were removed and the supernatant containing released acetylcholinesterase collected, which was then used for colorimetric exosomes detection through the ability of acetylcholinesterase to hydrolyze acetylcholine to release acetic acid. The resulting decreased solution pH was detected with phenol red indicator, with the sharp color transition conveniently by naked eye. Exosomes quantification was also achieved using the solution's absorption intensity ratio of 558 vs. 432 nm. The linear range was from 2.0 × 103 to 5.0 × 105 particles/μL, and the limit of detection and limit of quantification were 1.2 × 103 particles/μL and 2.2 × 103 particles/μL, respectively. In addition, this proposed strategy for exosomes detection showed a relative standard deviation of 3.1% and high recovery efficiency (>94%), exhibiting a bright application future in exsomes analysis.
Collapse
|
50
|
Xia Y, Chen T, Chen W, Chen G, Xu L, Zhang L, Zhang X, Sun W, Lan J, Lin X, Chen J. A dual-modal aptasensor based on a multifunctional acridone derivate for exosomes detection. Anal Chim Acta 2022; 1191:339279. [PMID: 35033266 DOI: 10.1016/j.aca.2021.339279] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/22/2021] [Accepted: 11/10/2021] [Indexed: 11/01/2022]
Abstract
Exosomes are promising biomarkers for cancer screening, but the development of a robust approach that can sensitively and accurately detect exosomes remains challenging. In the present study, an aptasensor based on the multifunctional signal probe 10-benzyl-2-amino-acridone (BAA) was developed for the colorimetric and photoelectrochemical detection and quantitation of exosomes. Exosomes are captured by cholesterol DNA anchor-modified magnetic beads (MBs) through hydrophobic interactions. This capture process can be monitored under a confocal fluorescence microscope using BAA as the fluorescent signal probe. The aptamer modified copper oxide nanoparticles (CuO NPs) then bind to mucin 1 (MUC1) on the surface of the exosomes to form a sandwich structure (MBs-Exo-CuO NPs). Finally, the MBs-Exo-CuO NPs are dissolved in nitric acid to generate Cu2+, which inhibits the visible-light-induced oxidase mimic activity and photoelectrochemical activity of BAA simultaneously. The changes in absorbance and photocurrent intensities are directly proportional to the concentration of exosomes. In this dual-modal aptasensor, the colorimetric assay can achieve rapid screening and identification, which is especially useful for point-of-care testing. The UV-vis absorbance and photocurrent assays then provide quantitative information, with a limit of detection of 1.09 × 103 particles μL-1 and 1.38 × 103 particles μL-1, respectively. The proposed aptasensor thus performs dual-modal detection and quantitation of exosomes. This aptasensor provides a much-needed toolset for exploring the biological roles of exosomes in specific diseases, particularly in the clinical setting.
Collapse
Affiliation(s)
- Yaokun Xia
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Tingting Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Wenqian Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Guanyu Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Lilan Xu
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Li Zhang
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Xiaoling Zhang
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Weiming Sun
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Jianming Lan
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China
| | - Xu Lin
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China.
| | - Jinghua Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian Province, 350108, PR China.
| |
Collapse
|