1
|
Ge W, Mu Z, Yang S, Zeng Y, Deng Y, Lin Y, Xie P, Li G. Biosensor-based methods for exosome detection with applications to disease diagnosis. Biosens Bioelectron 2025; 279:117362. [PMID: 40157151 DOI: 10.1016/j.bios.2025.117362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/09/2025] [Accepted: 03/09/2025] [Indexed: 04/01/2025]
Abstract
Exosomes are nanoscale extracellular vesicles (EVs) secreted by most eukaryotic cells and can be found in nearly all human body fluids. Increasing evidence has revealed their pivotal roles in intercellular communication, and their active participation in myriad physiological and pathological activities. Exosomes' functions rely on their contents that are closely correlated with the biological characteristics of parental cells, which may provide a rich resource of molecular information for accurate and detailed diagnosis of a diverse array of diseases, such as differential diagnosis of Alzheimer's disease, early detection and subtyping of various tumors. As a category of sensitive detection devices, biosensors can fully reveal the molecular information and convert them into actionable clinical information. In this review, recent advances in biosensor-based methods for the detection of exosomes are summarized. We have described the fabrication of various biosensors based on the analysis of exosomal proteins, RNAs or glycans for accurate diagnosis, with respect to their elaborate recognition designs, signal amplification strategies, sensing properties, as well as their application potential. The challenges along with corresponding technologies in the future development and clinical translation of these biosensors are also discussed.
Collapse
Affiliation(s)
- Weikang Ge
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Zheying Mu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Shiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yujing Zeng
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yifan Lin
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China.
| |
Collapse
|
2
|
Gu X, Fan Z, Lu L, Xu H, He L, Shen H, Huang R, Li Z. Machine learning-assisted washing-free detection of extracellular vesicles by target recycling amplification based fluorescent aptasensor for accurate diagnosis of gastric cancer. Talanta 2025; 287:127506. [PMID: 39837204 DOI: 10.1016/j.talanta.2024.127506] [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: 06/30/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/23/2025]
Abstract
Extracellular vesicles (EVs) are promising non-invasive biomarkers for cancer diagnosis. EVs proteins play a critical role in tumor progress and metastasis. However, accurately and reliably diagnosing cancers is greatly limited by single protein marker on EVs. Here, we reported an accurate diagnosis model of gastric cancer by analyzing five types of EVs surface proteins using machine learning in a retrospective study design. A washing-free detection method based on aptasensor and exonuclease Ⅰ was used to profile EVs surface proteins. The aptamer was designed as hairpin structure. The presence of target protein positive EVs converted the conformation of hairpin probes, which subsequently degraded by exonuclease Ⅰ. The exposed target protein could bind with and then open new hairpin probes, thus forming an amplification cycle. The lengths of different detection probes were optimized for detection. With the combination of five target proteins, five machine learning algorithms were compared to achieve a higher diagnostic accuracy. The best model, XGBoost, validated with 20 % of detection results could reach an accuracy of 0.8421. Furthermore, the XGBoost-based surface protein analysis could precisely identify gastric cancer patients with the area under the curve value of 0.9347 (95 % confidential interval (CI) = 0.8590 to 1.000). Since our method utilized a simple and versatile design of detection probes, its diagnostic scope could potentially be expanded to include different protein markers and accurately diagnose other diseases in the future.
Collapse
Affiliation(s)
- Xinrui Gu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Zeyu Fan
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China
| | - Luying Lu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Hongpan Xu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Lei He
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China.
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China.
| | - Zhiyang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| |
Collapse
|
3
|
Zhu A, Ahmad W, Xu Y, Wei W, Jiao T, Ouyang Q, Chen Q. Trace detection of S. aureus cells in food samples via RCA-assisted SERS signal amplification with core-shell nanoprobe. Talanta 2025; 286:127458. [PMID: 39755075 DOI: 10.1016/j.talanta.2024.127458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/18/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
Staphylococcus aureus (S. aureus) has been identified as a indicator of food contamination. In this study, a sensitive and accurate biosensor strategy for S. aureus through rolling circle amplification-assisted surface-enhanced Raman scattering (RCA-assisted-SERS), has been established. The work relies on the interaction between the aptamer and its partial complementary DNA strands fabricated on the surface of gold and silver-assisted magnetic microspheres and the subsequent detachment to trigger the activation of the RCA process. In RCA, template DNA, T4 DNA ligase, and Phi29 DNA polymerase were assembled to form long single-stranded DNA containing repetitive sequences. The gold core encapsulated with a layer of 4-nitrothiophenol and further covered with a silica shell was employed as the SERS nanoprobe (Au@NTP@SiO2). Subsequently, the output and amplification of SERS signal were performed by hybridizing ssDNA functionalized Au@NTP@SiO2 to realize the quantitative detection of S. aureus. Under the optimal conditions, S. aureus sensing was monitored (36.0-3.6 × 108 cfu/mL) with a limit of detection of 2.0 cfu/mL. This strategy was further validated for S. aureus recognition in spiked real samples with favorable recoveries (94.0-103.4 %) at p > 0.05. The suggested RCA-assisted SERS approach exhibits potential for multiple foodborne pathogens in both food safety and biomedical investigations.
Collapse
Affiliation(s)
- Afang Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Waqas Ahmad
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, PR China
| | - Yi Xu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, PR China
| | - Wenya Wei
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Tianhui Jiao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, PR China
| | - Qin Ouyang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China.
| | - Quansheng Chen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China; College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, PR China.
| |
Collapse
|
4
|
Zhang Q, Liu Q, Long K, Zhou K, Yang Z, Ge A, Hu J, Peng C, Wang W, Wang H, Li B. Visual and fluorescence dual mode platform for sensitive and accurate screening of breast carcinoma. Biosens Bioelectron 2025; 271:117047. [PMID: 39705784 DOI: 10.1016/j.bios.2024.117047] [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: 10/15/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024]
Abstract
Compared to single-mode detection, dual-mode sensing strategies have garnered increasing attention from researchers due to their superior detection accuracy and reliability. Exosomes, as non-invasive biomarkers, hold significant potential for disease diagnosis. However, sensitive and precise detection of exosomes still presents considerable technical challenges. Inspired by the advantages of dual-mode detection, we developed a visual and fluorescence dual-mode platform (VFDMP) based on an aptamer strategy for exosome detection using enzyme-free nucleic acid amplification and nanomaterial-assisted cation exchange reactions (CERs). The Aptamer-ssDNA complexes capture tumor-derived exosomes, releasing abundant single-stranded DNA (ssDNAs), which then triggers the catalytic hairpin assembly (CHA) cycle, leading to the release of Ag+. The introduced CdTe quantum dots (QDs) act as signal reporters, interacting with Ag+ through CERs, and switching both fluorescence and visual signals from "on" to "off" to achieve exosome detection. Based on this innovative sensing principle, the developed FL/visual dual-mode aptasensor demonstrated excellent sensitivity and accuracy, achieving a low detection limit of 1.1 particles/μL by fluorometer, while exosome concentrations as low as 300 particles/mL could be visually distinguished by naked eye. Furthermore, this dual-mode platform can directly detect exosomes in clinical human serum samples, with only a small volume (10 μL) required. It can accurately differentiate between healthy individuals and breast cancer patients, as well as identify cancer stages (Stage II and Stage III) and subtypes (triple-negative, luminal B, and HER2+). These results suggest that the developed dual-mode detection strategy holds great promise as a sensitive, accurate method for biomarker analysis in clinical samples.
Collapse
Affiliation(s)
- Qiongdan Zhang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Qingyi Liu
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Kang Long
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Kang Zhou
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Zheng Yang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Anqi Ge
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Jinhui Hu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Caiyun Peng
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China; Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China.
| | - Huizhen Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China.
| | - Bin Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China.
| |
Collapse
|
5
|
Li C, Xu T, Hou G, Wang Y, Fu Q. DNA nanotechnology-based strategies for gastric cancer diagnosis and therapy. Mater Today Bio 2025; 30:101459. [PMID: 39866794 PMCID: PMC11762204 DOI: 10.1016/j.mtbio.2025.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Gastric cancer (GC) is a formidable adversary in the field of oncology. The low early diagnosis rate of GC results in a low overall survival rate. Therefore, early accurate diagnosis and effective treatment are the key to reduce the mortality of GC. With the advent of nanotechnology, researchers continue to explore new possibilities for accurate diagnosis and effective treatment. One such breakthrough is the application of DNA nanotechnology. In this paper, the application of exciting DNA nanomaterials in the diagnosis and treatment of GC is discussed in depth. Firstly, the biomarkers related to GC and the diagnostic strategies related to DNA nanotechnology are summarized. Second, the latest research progress of DNA nanomaterials in the GC targeted therapy are summarized. Finally, the challenges and opportunities of DNA nanomaterials in the research and clinical application of GC are prospected.
Collapse
Affiliation(s)
- Congcong Li
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Tongyang Xu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Guopeng Hou
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Yin Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| | - Qinrui Fu
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266073, China
| |
Collapse
|
6
|
Hu J, Gao D. Recent Advances in Aptamer-Based Microfluidic Biosensors for the Isolation, Signal Amplification and Detection of Exosomes. SENSORS (BASEL, SWITZERLAND) 2025; 25:848. [PMID: 39943486 PMCID: PMC11820184 DOI: 10.3390/s25030848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025]
Abstract
Exosomes carry diverse tumor-associated molecular information that can reflect real-time tumor progression, making them a promising tool for liquid biopsy. However, traditional methods for exosome isolation and detection often rely on large, expensive equipment and are time-consuming, limiting their practical applicability in clinical settings. Microfluidic technology offers a versatile platform for exosome analysis, with advantages such as seamless integration, portability and reduced sample volumes. Aptamers, which are single-stranded oligonucleotides with high affinity and specificity for target molecules, have been frequently employed in the development of aptamer-based microfluidics for the isolation, signal amplification, and quantitative detection of exosomes. This review summarizes recent advances in aptamer-based microfluidic strategies for exosome analysis, including (1) strategies for on-chip exosome capture mediated by aptamers combined with nanomaterials or nanointerfaces; (2) aptamer-based on-chip signal amplification techniques, such as enzyme-free hybridization chain reaction (HCR), rolling circle amplification (RCA), and DNA machine-assisted amplification; and (3) various aptamer-assisted detection methods, such as fluorescence, electrochemistry, surface-enhanced Raman scattering (SERS), and magnetism. The limitations and advantages of these methods are also summarized. Finally, future challenges and directions for the clinical analysis of exosomes based on aptamer-based microfluidics are discussed.
Collapse
Affiliation(s)
- Jessica Hu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School and Open FIESTA, Tsinghua University, Shenzhen 518055, China;
- Key Laboratory of Metabolomics at Shenzhen, Shenzhen 518055, China
| | - Dan Gao
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School and Open FIESTA, Tsinghua University, Shenzhen 518055, China;
- Key Laboratory of Metabolomics at Shenzhen, Shenzhen 518055, China
| |
Collapse
|
7
|
Zhao S, Kong H, Qi D, Qiao Y, Li Y, Cao Z, Wang H, He X, Liu H, Yang H, Gao S, Liu T, Xie J. Epidermal stem cell derived exosomes-induced dedifferentiation of myofibroblasts inhibits scarring via the miR-203a-3p/PIK3CA axis. J Nanobiotechnology 2025; 23:56. [PMID: 39881312 PMCID: PMC11776291 DOI: 10.1186/s12951-025-03157-9] [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: 10/25/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Hypertrophic scar (HS) is a common fibroproliferative disorders with no fully effective treatments. The conversion of fibroblasts to myofibroblasts is known to play a critical role in HS formation, making it essential to identify molecules that promote myofibroblast dedifferentiation and to elucidate their underlying mechanisms. In this study, we used comparative transcriptomics and single-cell sequencing to identify key molecules and pathways that mediate fibrosis and myofibroblast transdifferentiation. Epidermal stem cell-derived extracellular vesicles (EpiSC-EVs) were isolated via ultracentrifugation and filtration, followed by miRNA sequencing to identify miRNAs targeting key molecules. After in vitro and in vivo treatment with EpiSC-EVs, we assessed antifibrotic effects through scratch assays, collagen contraction assays, Western blotting, and immunofluorescence. Transcriptomic sequencing and rescue experiments were used to investigate the molecular mechanism by which miR-203a-3p in EpiSC-EVs induces myofibroblast dedifferentiation. Our results indicate that PIK3CA is overexpressed in HS tissues and positively correlates with fibrosis. EpiSC-EVs were absorbed by scar-derived fibroblasts, promoting dedifferentiation from myofibroblasts to quiescent fibroblasts. Mechanistically, miR-203a-3p in EpiSC-EVs plays an essential role in inhibiting PIK3CA expression and PI3K/AKT/mTOR pathway hyperactivation, thereby reducing scar formation. In vivo studies confirmed that EpiSC-EVs attenuate excessive scarring through the miR-203a-3p/PIK3CA axis, suggesting EpiSC-EVs as a promising therapeutic approach for HS.
Collapse
Affiliation(s)
- Shixin Zhao
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Haoran Kong
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Dahu Qi
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Yushuang Qiao
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Yu Li
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Zhiming Cao
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China
| | - Hanwen Wang
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xuefeng He
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Hengdeng Liu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Hao Yang
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Suyue Gao
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Tao Liu
- Department of Traumatic Orthopedics, Henan Provincial People's Hospital & The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China.
- Henan Orthopedics Research Institute, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, China.
| | - Julin Xie
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
8
|
Li Y, Lv X, Jiang H, Li X, Deng Y. Integration of RCA-Based DNA Nanoscaffold with Target Triggered RNA-Cleaving DNAzyme for Sensitive Detection of miRNA21. Appl Biochem Biotechnol 2024; 196:8925-8939. [PMID: 39083195 DOI: 10.1007/s12010-024-05022-1] [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] [Accepted: 07/23/2024] [Indexed: 01/04/2025]
Abstract
Cascaded amplification showed promising potential for detection of trace target miRNAs in molecular diagnosis and prevention of many diseases. In this study, miRNA21 was chosen as the target, and rolling circle amplification (RCA)-based DNA nanoscaffold was integrated with target triggered RNA-cleaving DNAzyme for sensitive detection of miRNA21. That is, the H1 probe was bound with the long-chain product of RCA to self-assemble into DNA nanoscaffold. Target miRNA21 triggered the hybridization chain reaction (HCR) located on the nanoscaffold, and led to rapid proximity of DNAzyme fragments modified at both ends of the H2 probe, which realized the cyclic cleavage of self-quenching substrate probe efficiently, and the fluorescence signal was restored. The results demonstrated that the proposed assay was sensitive, 0.76 pM of miRNA21 can be detected. The proposed assay was specific; only one-base mismatched miRNA21 can be effectively recognized, other nucleic acid sequence and the serum matrix did not cause any interference. The proposed assay was accurate; recoveries from 82.1 to 115.0% can be obtained in the spiked fetal bovine serum (FBS). The flexible and programmable characteristics of DNA nanoscaffold and DNAzyme provide a confident and robust strategy for more sensitive nucleic acid detection, and can be developed to be a universal sensing platform for detecting other miRNAs just needing modification on the corresponding sequence of H1 probe in HCR.
Collapse
Affiliation(s)
- Yuan Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Xuefei Lv
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China.
| | - Hao Jiang
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Xiaoqiong Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yulin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
9
|
Zhang L, Bai H, Zou J, Zhang C, Zhuang W, Hu J, Yao Y, Hu WW. Immuno-Rolling Circle Amplification (Immuno-RCA): Biosensing Strategies, Practical Applications, and Future Perspectives. Adv Healthc Mater 2024; 13:e2402337. [PMID: 39252654 DOI: 10.1002/adhm.202402337] [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: 06/26/2024] [Revised: 08/25/2024] [Indexed: 09/11/2024]
Abstract
In the rapidly evolving field of life sciences and biomedicine, detecting low-abundance biomolecules, and ultraweak biosignals presents significant challenges. This has spurred a rapid development of analytical techniques aiming for increased sensitivity and specificity. These advancements, including signal amplification strategies and the integration of biorecognition events, mark a transformative era in bioanalytical precision and accuracy. A prominent method among these innovations is immuno-rolling circle amplification (immuno-RCA) technology, which effectively combines immunoassays with signal amplification via RCA. This process starts when a targeted biomolecule, such as a protein or cell, binds to an immobilized antibody or probe on a substrate. The introduction of a circular DNA template triggers RCA, leading to exponential amplification and significantly enhanced signal intensity, thus the target molecule is detectable and quantifiable even at the single-molecule level. This review provides an overview of the biosensing strategy and extensive practical applications of immuno-RCA in detecting biomarkers. Furthermore, it scrutinizes the limitations inherent to these sensors and sets forth expectations for their future trajectory. This review serves as a valuable reference for advancing immuno-RCA in various domains, such as diagnostics, biomarker discovery, and molecular imaging.
Collapse
Affiliation(s)
- Limei Zhang
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hao Bai
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jie Zou
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chuyan Zhang
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Weihua Zhuang
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jie Hu
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongchao Yao
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wenchuang Walter Hu
- Precision Medicine Translational Research Center (PMTRC), Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
10
|
Kuang J, Zhao L, Ruan S, Sun Y, Wu Z, Zhang H, Zhang M, Hu P. The integration platform for exosome capture and colorimetric detection: Site occupying effect-modulated MOF-aptamer interaction and aptamer-Au NPs-dopamine interaction. Anal Chim Acta 2024; 1329:343234. [PMID: 39396297 DOI: 10.1016/j.aca.2024.343234] [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: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024]
Abstract
Exosomes are extracellular vesicles of 30-200 nm in diameter that inherit molecular markers from their parent cells, including proteins, lipids, nucleic acids, and glycoconjugates. The detection and protein profiling of exosome could provide noninvasive access to disease diagnosis and treatment. In recent years, it has been found that Zr-MOFs can capture exosomes by forming Zr-O-P bonds through the phospholipid bilayer of exosomes. In addition, gold nanoparticles with optical response are used for colorimetric biological analysis, such as proteins, peptides, DNA. In this work, we proposed an aptasensor for exosome capture and sensitive colorimetric detection. The Zr-MOF (PCN-224) is innovatively used to capture exosome by Zr-O-P bond, and sodium tripolyphosphate (STPP) is used to block the non-specific adsorption of DNA aptamers on the surface of PCN-224 by site occupying effect. The aptamer binds to exosome immunity, and the remaining aptamer binds to Au NPs, resulting in an increase in steric hindrance and electrostatic repulsion, which makes the dispersion of Au NPs better and avoids the aggregation of Au NPs induced by dopamine (DA). The ratio of absorbance A650/A520 represents the aggregate degree of Au NPs, which correlates with the concentration of exosomes, and achieves sensitive colorimetric detection of exosomes with a linear range of 1.0 × 105-1.0 × 107 particles/mL. Further studies reveal that our work has excellent selectivity and anti-interference, breast cancer patients and healthy individuals can be distinguished by analyzing the differences in the expression of CD63 protein on exosome. The proposed biosensor integrates the capture and detection of exosomes, the multiple colors of Au NPs changed significantly from red to gray, which was conducive to the naked-eye identification of exosome detection.
Collapse
Affiliation(s)
- Jingjing Kuang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Linghao Zhao
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shengli Ruan
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yangkun Sun
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zeyu Wu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hongyang Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Min Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| | - Ping Hu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
11
|
Liang Z, Wang P, Li Z, Li W, Ma Q. Au Nanorings/TiO 2 NPs@MXene-Based Metasurfaces with a Magnetic Mirror-Modulated ECL Strategy for Extracellular Vesicle Detection. Anal Chem 2024; 96:16443-16452. [PMID: 39347690 DOI: 10.1021/acs.analchem.4c04460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
A metasurface as an artificial electromagnetic structure can concentrate optical energy into nanometric volumes to strongly enhance the light-matter interaction, which has been becoming a powerful platform for optical sensing, nonlinear effects, and quantum optics. Herein, we developed a novel hybrid plasmonic-dielectric metasurface consisting of Au nanorings (Au NRs) and TiO2 nanoparticles derived from MXene (TiO2 NPs@MXene). The hybrid metasurface simultaneously benefited from the high near-field enhancement effect of plasmonic materials and the low loss of dielectric materials. Furthermore, the optical modulation efficiency of the hybrid metasurface can be regulated by a magnetic mirror configuration. The magnetic mirror acted like a mirror, confining the electrons to a limited region and increasing the density of the surface plasmon. Moreover, the electrochemiluminescence (ECL) of the Cu2BDC metal-organic framework (Cu2BDC-MOF) served as a light source for the Au NRs/TiO2 NPs@MXene metasurface. Due to the exceptional light manipulation capability of the hybrid metasurface and the coordination of the magnetic mirror, the isotropic ECL signal can be dynamically amplified and converted into polarized emission. Finally, a metasurface-regulated ECL (MECL)-based biosensor with a dual-positive membrane protein recognition strategy was developed for the accurate identification of gastric cancer-derived extracellular vesicles. The novel MECL research opened up a new route in the realization of dynamically tunable metasurfaces for optical sensing and novel nanophotonic devices.
Collapse
Affiliation(s)
- Zihui Liang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Peilin Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhenrun Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Wenyan Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Qiang Ma
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
12
|
Tang H, Yu D, Zhang J, Wang M, Fu M, Qian Y, Zhang X, Ji R, Gu J, Zhang X. The new advance of exosome-based liquid biopsy for cancer diagnosis. J Nanobiotechnology 2024; 22:610. [PMID: 39380060 PMCID: PMC11463159 DOI: 10.1186/s12951-024-02863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Liquid biopsy is a minimally invasive method that uses biofluid samples instead of tissue samples for cancer diagnosis. Exosomes are small extracellular vesicles secreted by donor cells and act as mediators of intercellular communication in human health and disease. Due to their important roles, exosomes have been considered as promising biomarkers for liquid biopsy. However, traditional methods for exosome isolation and cargo detection methods are time-consuming and inefficient, limiting their practical application. In the past decades, many new strategies, such as microfluidic chips, nanowire arrays and electrochemical biosensors, have been proposed to achieve rapid, accurate and high-throughput detection and analysis of exosomes. In this review, we discussed about the new advance in exosome-based liquid biopsy technology, including isolation, enrichment, cargo detection and analysis approaches. The comparison of currently available methods is also included. Finally, we summarized the advantages and limitations of the present strategies and further gave a perspective to their future translational use.
Collapse
Affiliation(s)
- Haozhou Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
- Department of Orthopaedics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Dan Yu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jiahui Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Maoye Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Min Fu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Qian
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoxin Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Runbi Ji
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jianmei Gu
- Departmemt of Clinical Laboratory Medicine, Nantong Tumor Hospital/Affiliated Tumor Hospital of Nantong University, Nantong, 226300, China.
- Affiliated Cancer Hospital of Nantong University, Nantong, 226300, China.
| | - Xu Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
13
|
Yao X, He D, Wei P, Niu Z, Chen H, Li L, Fu P, Wang Y, Lou S, Qian S, Zheng J, Zuo G, Wang K. DNA Nanomaterial-Empowered Surface Engineering of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306852. [PMID: 38041689 DOI: 10.1002/adma.202306852] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/30/2023] [Indexed: 12/03/2023]
Abstract
Extracellular vesicles (EVs) are cell-secreted biological nanoparticles that are critical mediators of intercellular communication. They contain diverse bioactive components, which are promising diagnostic biomarkers and therapeutic agents. Their nanosized membrane-bound structures and innate ability to transport functional cargo across major biological barriers make them promising candidates as drug delivery vehicles. However, the complex biology and heterogeneity of EVs pose significant challenges for their controlled and actionable applications in diagnostics and therapeutics. Recently, DNA molecules with high biocompatibility emerge as excellent functional blocks for surface engineering of EVs. The robust Watson-Crick base pairing of DNA molecules and the resulting programmable DNA nanomaterials provide the EV surface with precise structural customization and adjustable physical and chemical properties, creating unprecedented opportunities for EV biomedical applications. This review focuses on the recent advances in the utilization of programmable DNA to engineer EV surfaces. The biology, function, and biomedical applications of EVs are summarized and the state-of-the-art achievements in EV isolation, analysis, and delivery based on DNA nanomaterials are introduced. Finally, the challenges and new frontiers in EV engineering are discussed.
Collapse
Affiliation(s)
- Xuxiang Yao
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Dongdong He
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Pengyao Wei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Zitong Niu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Hao Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lin Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Yiting Wang
- College of Chemistry, Jilin Normal University, Siping, 136000, P. R. China
| | - Saiyun Lou
- Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
- Ningbo Second Hospital, Ningbo, 315010, P. R. China
| | - Sihua Qian
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Jianping Zheng
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Guokun Zuo
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| |
Collapse
|
14
|
Mazahir F, Yadav AK. Recent progress in engineered extracellular vesicles and their biomedical applications. Life Sci 2024; 350:122747. [PMID: 38797364 DOI: 10.1016/j.lfs.2024.122747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
AIMS To present the recent update on the isolation, engineering techniques for extracellular vesicles, limitations associated with different isolation techniques, different biomedical applications, and challenges of engineered extracellular vesicles for the benefit of researchers from academic, industry, etc. MATERIALS AND METHODS: Peer-reviewed articles from most recognized journals were collected, and presented information was analyzed to discuss collection, chemical, electroporation, cellular, and membrane surface engineering to design extracellular vesicles for various therapeutic applications. In addition, we present the applications and limitations of techniques for the collection of extracellular vesicles. KEY FINDINGS There is a need for isolation techniques with the gold standard. However, advanced extracellular vesicle isolation techniques showed improved recovery, and purity of extracellular vesicles. Tumor therapy is a major part of the therapy section that illustrates the role of engineered extracellular vesicles in synergetic therapy such as phototherapy, theragnostic, and delivery of genetic materials. In addition, extracellular vesicles have shown their potential in the treatment of retinal disorders, neurodegenerative disease, tuberculosis, osteoporosis, inflammatory bowel disease, vaccine production, and wound healing. SIGNIFICANCE Engineered extracellular vesicles can deliver cargo to the specific cells, elicit an immune response and could be used for the development of the vaccines in the future. However, the progress is at the initial stage. Overall, this review will provide a comprehensive understanding and could serve as a reference for researchers in the clinical translation of engineered extracellular vesicles in different biomedical fields.
Collapse
Affiliation(s)
- Farhan Mazahir
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India.
| |
Collapse
|
15
|
Cai D, Chen GL, Wang T, Zhang KH. Trends and frontiers in signal amplification for aptamer-based tumor detection: A bibliometric analysis. World J Clin Cases 2024; 12:4726-4741. [PMID: 39070802 PMCID: PMC11235479 DOI: 10.12998/wjcc.v12.i21.4726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Malignant tumors are one of the leading causes of death worldwide, imposing a substantial economic and social burden. Early detection is the key to improving cure rates and reducing mortality rates, which requires the development of sensitive early detection technologies. Signal amplification techniques play a crucial role in aptamer-based early detection of tumors and are increasingly garnering attention from researchers. AIM To investigate the current research status, developmental trajectories, and hotspots in signal amplification for aptamer-based tumor detection through bibliometric analysis. METHODS English publications pertaining to signal amplification in aptamer-based tumor detection were retrieved from the Web of Science Core Collection database. VOSviewer and CiteSpace software were employed to analyze various information within this field, including countries, institutions, authors, co-cited authors, journals, co-cited journals, cited references, and keywords. RESULTS A total of 757 publications were included in this study. China accounted for 85.47% of all publications, with Nanjing University (China) emerging as the institution with the highest publication output. The most influential authors and journals were Hasanzadeh M. from Iran and "Biosensors and Bioelectronics", respectively. Exosomes and carcinoembryonic antigen (CEA) stood out as the most researched tumor-related molecules. Currently, the predominant signal amplification technique, nanomaterial, and signal transduction method were identified as hybridization chain reactions, gold nanoparticles, and electrochemical methods, respectively. Over the past 3 years, exosomes, CEA, electrochemical biosensors, and nanosheets have emerged as research hotspots, exhibiting a robust burst of intensity. CONCLUSION This study is the first bibliometric analysis of literature on signal amplification in aptamer-based tumor detection and elucidates the current status, hotspots, and prospective research directions within this realm. Additionally, it provides an important reference for researchers.
Collapse
Affiliation(s)
- Dan Cai
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Jiangxi Institute of Gastroenterology and Hepatology, Nanchang 330006, Jiangxi Province, China
| | - Gui-Lin Chen
- Department of Anorectal Surgery, The 908th Hospital of the Chinese People's Liberation Army Joint Logistics Support Force, Nanchang 330000, Jiangxi Province, China
| | - Ting Wang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Jiangxi Institute of Gastroenterology and Hepatology, Nanchang 330006, Jiangxi Province, China
| | - Kun-He Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Jiangxi Institute of Gastroenterology and Hepatology, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
16
|
Wu N, Wong KY, Yu X, Zhao JW, Zhang XY, Wang JH, Yang T. Multispectral 3D DNA Machine Combined with Multimodal Machine Learning for Noninvasive Precise Diagnosis of Bladder Cancer. Anal Chem 2024; 96:10046-10055. [PMID: 38845359 DOI: 10.1021/acs.analchem.4c01749] [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: 06/19/2024]
Abstract
Extracellular vesicle (EV) molecular phenotyping offers enormous opportunities for cancer diagnostics. However, the majority of the associated studies adopted biomarker-based unimodal analysis to achieve cancer diagnosis, which has high false positives and low precision. Herein, we report a multimodal platform for the high-precision diagnosis of bladder cancer (BCa) through a multispectral 3D DNA machine in combination with a multimodal machine learning (ML) algorithm. The DNA machine was constructed using magnetic microparticles (MNPs) functionalized with aptamers that specifically identify the target of interest, i.e., five protein markers on bladder-cancer-derived urinary EVs (uEVs). The aptamers were hybridized with DNA-stabilized silver nanoclusters (DNA/AgNCs) and a G-quadruplex/hemin complex to form a sensing module. Such a DNA machine ensured multispectral detection of protein markers by fluorescence (FL), inductively coupled plasma mass spectrometry (ICP-MS), and UV-vis absorption (Abs). The obtained data sets then underwent uni- or multimodal ML for BCa diagnosis to compare the analytical performance. In this study, urine samples were obtained from our prospective cohort (n = 45). Our analytical results showed that the 3D DNA machine provided a detection limit of 9.2 × 103 particles mL-1 with a linear range of 4 × 104 to 5 × 107 particles mL-1 for uEVs. Moreover, the multimodal data fusion model exhibited an accuracy of 95.0%, a precision of 93.1%, and a recall rate of 93.2% on average, while those of the three types of unimodal models were no more than 91%. The elevated diagnosis precision by using the present fusion platform offers a perspective approach to diminishing the rate of misdiagnosis and overtreatment of BCa.
Collapse
Affiliation(s)
- Na Wu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
- Institute of Precision Medicine, Fujian Medical University, Fuzhou 350122, China
| | - Ka-Ying Wong
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Xin Yu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Jia-Wei Zhao
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Xin-Yu Zhang
- General Hospital of Northern Theater Command, Shenyang, Liaoning 110015, China
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Ting Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| |
Collapse
|
17
|
Zheng LE, Huang M, Liu Y, Bao Q, Huang Y, Ye Y, Liu M, Sun P. Colorimetric aptasensor based on temporally controllable light-stimulated oxidase-mimicking fluorescein for the sensitive detection of exosomes in mild conditions. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3577-3586. [PMID: 38787692 DOI: 10.1039/d4ay00561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Analysis of exosomes provides important information for rapid and non-invasive screening of tumors. However, sensitive and convenient detection of exosomes remains technically challenging to date. Herein, a colorimetric aptasensor based on the light-stimulated oxidase-mimicking activity of FITC was constructed for detecting ovarian cancer (OC) exosomes. The aptasensor contained an EpCAM aptamer to capture OC exosomes. Cholesterol and fluorescein (FITC) were used to modify either end of the DNA (DNA anchor). The DNA anchor could combine with exosomes through a hydrophobic reaction between cholesterol and the lipid membrane. FITC oxidized 3,3',5,5'-tetramethylbenzidine (TMB) under a 365 nm LED light source in a temporally controllable manner under mild conditions, causing the solution to change from colorless to blue, and the corresponding UV-vis absorbance increased. Based on this principle, the exosomes were qualitatively analyzed by observing the color change with the naked eye. In parallel, the exosome concentration was also detected using UV-vis spectrophotometry. The linear range was from 2 × 105 to 100 × 105 particles per mL with a limit of detection of 1.77 × 105 particles per mL. The developed aptasensor also exhibited favorable selectivity and could discriminate the exosomes from OC cells and normal cells. Besides, the receiver operating characteristic (ROC) curve demonstrates that it is possible to distinguish between patients with OC and healthy donors (HDs) using exosomes as the biomarker. Our technology may expand the applications of DNA-based detection method-enabled OC diagnostic tools.
Collapse
Affiliation(s)
- Li-E Zheng
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Min Huang
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yiyang Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Qiufang Bao
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yuxiu Huang
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yuhong Ye
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Mengmeng Liu
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China.
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, Fujian, China
| |
Collapse
|
18
|
Du S, Pei X, Huang Y, Wang Y, Li Z, Niu X, Zhang W, Sun W. Hemin/G-quadruplex and AuNPs-MoS 2 based novel dual signal amplification strategy for ultrasensitively sandwich-type electrochemical thrombin aptasensor. Bioelectrochemistry 2024; 157:108635. [PMID: 38185025 DOI: 10.1016/j.bioelechem.2023.108635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/09/2024]
Abstract
In this work, a novel sandwich-type electrochemical aptasensor based on the dual signal amplification strategy of hemin/G-quadruplex and AuNPs-MoS2 was designed and constructed, which realized the highly sensitive and specific detection of thrombin (TB). In this aptasensor, the 15-mer TB-binding aptamer (TBA-1) modified with thiol group was immobilized on the surface of AuNPs modified glassy carbon electrode (AuNPs/GCE) as capturing elements. Another thiol-modified 29-mer TB-binding aptamer (TBA-2) sequence containing G-quadruplex structure for hemin immobilization was designed. The formed hemin/G-quadruplex/TBA-2 sequence was further combined to the AuNPs decorated flower-like molybdenum disulfide (AuNPs-MoS2) composite surface via Au-S bonds, acting the role of reporter probe. In presence of the target TB, the sandwich-type electrochemical aptamer detection system could be formed properly. With the assistance of the dual signal amplification of AuNPs-MoS2 and hemin/G-quadruplex toward H2O2 reduction, the sandwich-type electrochemical aptasensor was successfully constructed for sensitive detection of TB. The results demonstrate that the fabricated aptasensor displays a wide linear range of 1.0 × 10-6 ∼ 10.0 nM with a low detection limit of 0.34 fM. This proposed aptasensor shows potential application in the detection of TB content in real biological samples with high sensitivity, selectivity, and reliability.
Collapse
Affiliation(s)
- Shina Du
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Xiaoying Pei
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Yan Huang
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Yuebo Wang
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Zhongfang Li
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Xueliang Niu
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China.
| | - Weili Zhang
- College of Pharmacy, Key Laboratory of Biomedical Engineering and Technology in Universities of Shandong, Qilu Medical University, Zibo 255300, PR China.
| | - Wei Sun
- Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, PR China
| |
Collapse
|
19
|
Chen J, Zheng M, Xiao Q, Wang H, Chi C, Lin T, Wang Y, Yi X, Zhu L. Recent Advances in Microfluidic-Based Extracellular Vesicle Analysis. MICROMACHINES 2024; 15:630. [PMID: 38793203 PMCID: PMC11122811 DOI: 10.3390/mi15050630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
Extracellular vesicles (EVs) serve as vital messengers, facilitating communication between cells, and exhibit tremendous potential in the diagnosis and treatment of diseases. However, conventional EV isolation methods are labor-intensive, and they harvest EVs with low purity and compromised recovery. In addition, the drawbacks, such as the limited sensitivity and specificity of traditional EV analysis methods, hinder the application of EVs in clinical use. Therefore, it is urgent to develop effective and standardized methods for isolating and detecting EVs. Microfluidics technology is a powerful and rapidly developing technology that has been introduced as a potential solution for the above bottlenecks. It holds the advantages of high integration, short analysis time, and low consumption of samples and reagents. In this review, we summarize the traditional techniques alongside microfluidic-based methodologies for the isolation and detection of EVs. We emphasize the distinct advantages of microfluidic technology in enhancing the capture efficiency and precise targeting of extracellular vesicles (EVs). We also explore its analytical role in targeted detection. Furthermore, this review highlights the transformative impact of microfluidic technology on EV analysis, with the potential to achieve automated and high-throughput EV detection in clinical samples.
Collapse
Affiliation(s)
- Jiming Chen
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Meiyu Zheng
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Qiaoling Xiao
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Hui Wang
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Caixing Chi
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Tahui Lin
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Yulin Wang
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Xue Yi
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| | - Lin Zhu
- Department of Basic Medicine, Xiamen Medical College, Xiamen 361023, China; (J.C.); (M.Z.); (Q.X.); (H.W.); (C.C.); (T.L.); (Y.W.)
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen 361023, China
- Institute of Respiratory Diseases, Xiamen Medical College, Xiamen 361023, China
| |
Collapse
|
20
|
Deng J, Liu C, Sun J. DNA-Based Nanomaterials for Analysis of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303092. [PMID: 38016069 DOI: 10.1002/adma.202303092] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/21/2023] [Indexed: 11/30/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanovesicles comprising a myriad of molecular cargo such as proteins and nucleic acids, playing essential roles in intercellular communication and physiological and pathological processes. EVs have received substantial attention as noninvasive biomarkers for disease diagnosis and prognosis. Owing to their ability to recognize protein and nucleic acid targets, DNA-based nanomaterials with excellent programmability and modifiability provide a promising tool for the sensitive and accurate detection of molecular cargo carried by EVs. In this perspective, recent advancements in EV analysis using a variety of DNA-based nanomaterials are summarized, which can be broadly classified into three categories: linear DNA probes, DNA nanostructures, and hybrid DNA nanomaterials. The design, construction, advantages, and disadvantages of different types of DNA nanomaterials, as well as their performance for detecting EVs are reviewed. The challenges and opportunities in the field of EV analysis by DNA nanomaterials are also discussed.
Collapse
Affiliation(s)
- Jinqi Deng
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Liu
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiashu Sun
- Beijing Engineering Research Center for BioNanotechnology, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
21
|
Mo F, Lin C, Lu J, Sun D. Integrating Artificial DNAzymes with Natural Enzymes on 2D MOF Hybrid Nanozymes for Enhanced Treatment of Bacteria-Infected Wounds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307256. [PMID: 38018326 DOI: 10.1002/smll.202307256] [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: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Removal of invasive bacteria is critical for proper wound healing. This task is challenging because these bacteria can trigger intense oxidative stress and gradually develop antibiotic resistance. Here, the use of a multienzyme-integrated nanocatalytic platform is reported for efficient bacterial clearance and mitigation of inflammatory responses, constructed by physically adsorbing natural superoxide dismutase (SOD), in situ reduction of gold nanoparticles (Au NPs), and incorporation of a DNAzyme on 2D NiCoCu metal-organic frameworks (DNAzyme/SOD/Au@NiCoCu MOFs, termed DSAM), which can adapt to infected wounds. O2 and H2O2 replenishment is achieved and alleviated the hypoxic microenvironment using the antioxidant properties of SOD. The H2O2 produced during the reaction is decomposed by peroxidase (POD)-like activity enhanced by Au NPs and DNAzyme, releasing highly toxic hydroxyl radicals (•OH) to kill the bacteria. In addition, it possesses glutathione peroxidase (GPx)-like activity, which depletes GSH and prevents •OH loss. Systematic antimicrobial tests are performed against bacteria using this multienzyme-integrated nanoplatform. A dual-mode strategy involving natural enzyme-enhanced antioxidant capacity and artificial enzyme-enhanced •OH release to develop an efficient and novel enzyme-integrated therapeutic platform is integrated.
Collapse
Affiliation(s)
- Fayin Mo
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Specialty of Clinical Pharmacy, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, China
| | - Chuyan Lin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Jing Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Duanping Sun
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Specialty of Clinical Pharmacy, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510699, China
| |
Collapse
|
22
|
Javed A, Kong N, Mathesh M, Duan W, Yang W. Nanoarchitectonics-based electrochemical aptasensors for highly efficient exosome detection. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2345041. [PMID: 38742153 PMCID: PMC11089931 DOI: 10.1080/14686996.2024.2345041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
Exosomes, a type of extracellular vesicles, have attracted considerable attention due to their ability to provide valuable insights into the pathophysiological microenvironment of the cells from which they originate. This characteristic implicates their potential use as diagnostic disease biomarkers clinically, including cancer, infectious diseases, neurodegenerative disorders, and cardiovascular diseases. Aptasensors, which are electrochemical aptamers based biosensing devices, have emerged as a new class of powerful detection technology to conventional methods like ELISA and Western analysis, primarily because of their capability for high-performance bioanalysis. This review covers the current research landscape on the detection of exosomes utilizing nanoarchitectonics strategy for the development of electrochemical aptasensors. Strategies involving signal amplification and biofouling prevention are discussed, with an emphasis on nanoarchitectonics-based bio-interfaces, showcasing their potential to enhance sensitivity and selectivity through optimal conduction and mass transport properties. The ongoing challenges to broaden the clinical applications of these biosensors are also highlighted.
Collapse
Affiliation(s)
- Aisha Javed
- School of Life and Environmental Science, Centre for Sustainable Bioproducts, Deakin University, Geelong, VIC, Australia
| | - Na Kong
- School of Life and Environmental Science, Centre for Sustainable Bioproducts, Deakin University, Geelong, VIC, Australia
| | - Motilal Mathesh
- School of Life and Environmental Science, Centre for Sustainable Bioproducts, Deakin University, Geelong, VIC, Australia
| | - Wei Duan
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC, Australia
| | - Wenrong Yang
- School of Life and Environmental Science, Centre for Sustainable Bioproducts, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
23
|
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
|
24
|
Lai R, Zeng X, Xu Q, Xu Y, Li X, Ru Y, Wang Y, Wang D, Zhou X, Shao Y. Ratiometric G-quadruplex/hemin DNAzymes with low-dosage associative substrates. Anal Chim Acta 2024; 1295:342320. [PMID: 38355221 DOI: 10.1016/j.aca.2024.342320] [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: 01/02/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND G-quadruplex (G4)/hemin DNAzymes with conversion of substrates into colorimetric readouts are well recognized as convenient biocatalysis tools in sensor development. However, the previously developed colorimetric G4/hemin DNAzymes are diffusive substrate-based DNAzymes (DSBDs). The current colorimetric DSBDs have several drawbacks including high dosage (∼mM) of diffusive substrates (DSs), colorimetric product toxicity, and single colorimetric readout without tolerance to fluctuation of experimental factors and background. In addition, the usage of high-dosage DSs can smear the G4 foldings and their discard is more harmful to environment. Therefore, exploring alternative DNAzymes with potential to overcome these drawbacks of DSBDs is urgently needed. RESULTS We herein developed associative substrate-based DNAzymes (ASBDs). Cyanine dyes were selected as associative substrates (ASs) due to their binding competency with G4/hemin DNAzymes. With respect to DSBDs, ASBDs needed only low dosage (∼10 μM) of ASs to be able to cause a rapid and visible substrate conversion. In addition, since cyanine dyes are NIR dyes with high extinction coefficients and their conversion products have absorption bands at shorter wavelength. Therefore, a colorimetric ratio response can be developed to follow activities of G4/hemin DNAzymes with competency to tolerate fluctuation of experimental factors and background. In particular, herein developed ASBDs can endure somewhat concentration fluctuation of H2O2. ASBDs are able to cowork with other enzymes (for example, glucose oxidase) to realize cascade sensing. SIGNIFICANCE The developed ASBDs can operate at low dosage of substrates with a colorimetric ratio response and can overcome the drawbacks met in DSBDs. We expect that, by designing ASs with fruitful color panel in the future, our work will inspire more interesting in developing environment-benign and low-carbon G4/hemin DNAzymes and desired colorful high-performance sensors.
Collapse
Affiliation(s)
- Rong Lai
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Xingli Zeng
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Qiuda Xu
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Ying Xu
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Xueni Li
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Yulu Ru
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Yilin Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Dandan Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Xiaoshun Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Yong Shao
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China.
| |
Collapse
|
25
|
Li Y, Shao F, Wu J, Liu M, Cao G, Zhao Z, Bai J, Gao Z. Ultrasensitive Ochratoxin A Detection in Cereal Products Using a Fluorescent Aptasensor Based on RecJ f Exonuclease-Assisted Target Recycling. Foods 2024; 13:595. [PMID: 38397572 PMCID: PMC10888426 DOI: 10.3390/foods13040595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Ochratoxin A (OTA) is a mycotoxin widely found in foodstuffs such as cereal grains. It greatly threatens human health owing to its strong toxicity and high stability. Aptasensors have emerged as promising tools for the analysis of small molecule contaminants. Nucleic-acid-based signal amplification enables detectable signals to be obtained from aptasensors. However, this strategy often requires the use of complex primers or multiple enzymes, entailing problems such as complex system instability. Herein, we propose a fluorescent aptasensor for the ultrasensitive detection of OTA in cereal products, with signal amplification through RecJf exonuclease-assisted target recycling. The aptamer/fluorescein-labeled complementary DNA (cDNA-FAM) duplex was effectively used as the target-recognition unit as well as the potential substrate for RecJf exonuclease cleavage. When the target invaded the aptamer-cDNA-FAM duplex to release cDNA-FAM, RecJf exonuclease could cleave the aptamer bonded with the target and release the target. Thus, the target-triggered cleavage cycling would continuously generate cDNA-FAM as a signaling group, specifically amplifying the response signal. The proposed exonuclease-assisted fluorescent aptasensor exhibited a good linear relationship with OTA concentration in the range from 1 pg/mL to 10 ng/mL with an ultralow limit of detection (6.2 ng/kg of cereal). The analytical method showed that recoveries of the cereal samples ranged from 83.7 to 109.3% with a repeatability relative standard deviation below 8%. Importantly, the proposed strategy is expected to become a common detection model because it can be adapted for other targets by replacing the aptamer. Thus, this model can guide the development of facile approaches for point-of-care testing applications.
Collapse
Affiliation(s)
- Yanxuan Li
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; (Y.L.); (F.S.); (J.W.); (M.L.); (J.B.); (Z.G.)
| | - Furong Shao
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; (Y.L.); (F.S.); (J.W.); (M.L.); (J.B.); (Z.G.)
- Department of Public Health and Management, Binzhou Medical University, Yantai 264003, China;
| | - Jin Wu
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; (Y.L.); (F.S.); (J.W.); (M.L.); (J.B.); (Z.G.)
| | - Mingzhu Liu
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; (Y.L.); (F.S.); (J.W.); (M.L.); (J.B.); (Z.G.)
| | - Gaofang Cao
- Department of Public Health and Management, Binzhou Medical University, Yantai 264003, China;
| | - Zunquan Zhao
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; (Y.L.); (F.S.); (J.W.); (M.L.); (J.B.); (Z.G.)
| | - Jialei Bai
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; (Y.L.); (F.S.); (J.W.); (M.L.); (J.B.); (Z.G.)
| | - Zhixian Gao
- Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; (Y.L.); (F.S.); (J.W.); (M.L.); (J.B.); (Z.G.)
- Department of Public Health and Management, Binzhou Medical University, Yantai 264003, China;
| |
Collapse
|
26
|
Zhao Z, Yang S, Tang X, Feng L, Ding Z, Chen Z, Luo X, Deng R, Sheng J, Xie S, Chang K, Chen M. DNA four-way junction-driven dual-rolling circle amplification sandwich-type aptasensor for ultra-sensitive and specific detection of tumor-derived exosomes. Biosens Bioelectron 2024; 246:115841. [PMID: 38006701 DOI: 10.1016/j.bios.2023.115841] [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: 10/17/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/27/2023]
Abstract
There is an urgent need to accurately quantify tumor-derived exosomes, which have emerged as promising non-invasive tumor diagnostic biomarkers. Herein, a bispecific-aptamer sandwich-type gold nanoparticle-modified electrochemical aptasensor was developed based on a four-way junction (4-WJ)-triggered dual rolling circle amplification (RCA)-assisted methylene blue (MB)/G-quadruplex strategy for extremely specific and sensitive exosome detection. This aptamer/exosome/aptamer sandwich-type design contained a CD63-specific aptamer and a cancerous mucin-1 (MUC1) protein-specific aptamer. The CD63 aptamer modified on a gold electrode captured exosomes, and then the sandwich-type aptasensor was formed with the addition of the MUC1 aptamer. The MUC1 aptamer's 3'-end sequence facilitated the formation of 4-WJ, assisted by a molecular beacon probe and a binary DNA probe. Subsequently, a dual-RCA reaction was triggered by binding to two cytosine-rich circle DNA templates at both ends of 4-WJ. Ultimately, dual-RCA products containing multiple G-quadruplex conformations were generated with the assistance of K+ to trap abundant MB indicators and amplify electrochemical signals. The aptasensor exhibited high specificity, sensitivity, repeatability, and stability toward MCF-7-derived exosomes, with a detection limit of 20 particles/mL and a linear range of 1 × 102 to 1 × 107 particles/mL. Moreover, it showed excellent applicability in clinical settings to recover exosomes in normal human serum. Our aptasensor is anticipated to serve as a versatile platform for detecting various specific aptamer-based targets in biomedical and bioanalytical applications.
Collapse
Affiliation(s)
- Zhuyang Zhao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Sha Yang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Liu Feng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Zishan Ding
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Zhiguo Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xing Luo
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Ruijia Deng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Jing Sheng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Shuang Xie
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China; College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.
| |
Collapse
|
27
|
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
|
28
|
Cheng S, Zhang C, Hu X, Zhu Y, Shi H, Tan W, Luo X, Xian Y. Ultrasensitive determination of surface proteins on tumor-derived small extracellular vesicles for breast cancer identification based on lanthanide-activated signal amplification strategy. Talanta 2024; 267:125189. [PMID: 37714039 DOI: 10.1016/j.talanta.2023.125189] [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: 05/16/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Small extracellular vesicles (sEVs) carrying multiple tumor-associated proteins inherited from parental cells play crucial roles in noninvasive breast cancer (BC) diagnosis. However, it is challenging to assess the subtle variations of surface proteins on sEV membranes due to the highly heterogeneous BC. Therefore, a simple and ultrasensitive assay based on lanthanide (Ln3+)-activated luminescence signal amplification was developed to detect multiple surface proteins on BC-derived sEVs. Multiple protein biomarkers on sEVs can be well identified with high sensitivity and specificity through dissolution-amplified luminescence of the NaEuF4 nanoparticle-based nanoprobe. We employ linear discriminant analysis to successfully discriminate triple negative BC cell (MDA-MB-231 cell) derived sEVs from other breast cell lines (MCF-7, SK-BR-3, BT474 and MCF-10A cell). Furthermore, the strategy enables high accuracy for districting the progression stages of BC patients and healthy donors. The simple and sensitive signal amplification strategy exhibits great potential for early clinic diagnosis by precise protein profiling of sEVs.
Collapse
Affiliation(s)
- Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| | - Xinyu Hu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yingxin Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenqiao Tan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Xianzhu Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
29
|
Xu H, Wu X, Liu Q, Yang C, Shen M, Wang Y, Liu S, Zhao S, Xiao T, Sun M, Ding Z, Bao J, Chen M, Gao M. A Universal Strategy for Enhancing the Circulating miRNAs' Detection Performance of Rolling Circle Amplification by Using a Dual-Terminal Stem-Loop Padlock. ACS NANO 2024; 18:436-450. [PMID: 38149638 PMCID: PMC10786163 DOI: 10.1021/acsnano.3c07721] [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: 08/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Rolling circle amplification (RCA) is one of the most promising nucleic acid detection technologies and has been widely used in the molecular diagnosis of disease. Padlock probes are often used to form circular templates, which are the core of RCA. However, RCA often suffers from insufficient specificity and sensitivity. Here we report a reconstruction strategy for conventional padlock probes to promote their overall performance in nucleic acid detection while maintaining probe functions uncompromised. When two rationally designed stem-loops were strategically placed at the two terminals of linear padlock probes, the specificity of target recognition was enhanced and the negative signal was significantly delayed. Our design achieved the best single-base discrimination compared with other structures and over a 1000-fold higher sensitivity than that of the conventional padlock probe, validating the effectiveness of this reconstruction. In addition, the underlying mechanisms of our design were elucidated through molecular dynamics simulations, and the versatility was validated with longer and shorter padlocks targeting the same target, as well as five additional targets (four miRNAs and dengue virus - 2 RNA mimic (DENV-2)). Finally, clinical applicability in multiplex detection was demonstrated by testing real plasma samples. Our exploration of the structures of nucleic acids provided another perspective for developing high-performance detection systems, improving the efficacy of practical detection strategies, and advancing clinical diagnostic research.
Collapse
Affiliation(s)
- Hanqing Xu
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Xianlan Wu
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Qian Liu
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Cheng Yang
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Man Shen
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Yingran Wang
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Shuai Liu
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Shuang Zhao
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Ting Xiao
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Minghui Sun
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Zishan Ding
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Jing Bao
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Ming Chen
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
- College
of Pharmacy and Laboratory Medicine, Third
Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing 400038, P. R. China
| | - Mingxuan Gao
- Department
of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| |
Collapse
|
30
|
Doherty C, Wilbanks B, Khatua S, Maher LJ. Aptamers in neuro-oncology: An emerging therapeutic modality. Neuro Oncol 2024; 26:38-54. [PMID: 37619244 PMCID: PMC10768989 DOI: 10.1093/neuonc/noad156] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Indexed: 08/26/2023] Open
Abstract
Despite recent advances in the understanding of brain tumor pathophysiology, challenges associated with tumor location and characteristics have prevented significant improvement in neuro-oncology therapies. Aptamers are short, single-stranded DNA or RNA oligonucleotides that fold into sequence-specific, 3-dimensional shapes that, like protein antibodies, interact with targeted ligands with high affinity and specificity. Aptamer technology has recently been applied to neuro-oncology as a potential approach to innovative therapy. Preclinical research has demonstrated the ability of aptamers to overcome some obstacles that have traditionally rendered neuro-oncology therapies ineffective. Potential aptamer advantages include their small size, ability in some cases to penetrate the blood-brain barrier, inherent lack of immunogenicity, and applicability for discovering novel biomarkers. Herein, we review recent reports of aptamer applications in neuro-oncology including aptamers found by cell- and in vivo- Systematic Evolution of Ligands by Exponential Enrichment approaches, aptamer-targeted therapeutic delivery modalities, and aptamers in diagnostics and imaging. We further identify crucial future directions for the field that will be important to advance aptamer-based drugs or tools to clinical application in neuro-oncology.
Collapse
Affiliation(s)
- Caroline Doherty
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Medical Scientist Training Program, Mayo Clinic Graduate School of Biomedical Sciences and Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Brandon Wilbanks
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
- Biochemistry and Molecular Biology Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Section of Neuro-Oncology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Louis James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| |
Collapse
|
31
|
Zou R, Cao L, Wu N, Chang G, Li L, Xiao L, Yan H, Li H, Wang P, Bao T, Zhang X, Wang S, Wang Y, He H. Transistor-based immunosensor using AuNPs-Ab2-HRP enzyme nanoprobe for the detection of antigen biomarker in human blood. Anal Bioanal Chem 2024; 416:163-173. [PMID: 37930375 DOI: 10.1007/s00216-023-05002-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023]
Abstract
Alpha-fetoprotein (AFP) is inextricably linked to various diseases, including liver cancer. Thus, detecting the content of AFP in biology has great significance in diagnosis, treatment, and intervention. Motivated by the urgent need for affordable and convenient electronic sensors in the analysis and detection of aqueous biological samples, we combined the solution-gated graphene transistor (SGGT) with the catalytic reaction of enzyme nanoprobes (HRP-AuNPs-Ab2) to accurately sense AFP. The SGGT immunosensor demonstrated high specificity and stability, excellent selectivity, and excessive linearity over a range of 4 ng/mL to 500 ng/mL, with the lower detection limit down to 1.03 ng/mL. Finally, clinical samples were successfully detected by the SGGT immunosensor, and the results were consistent with chemiluminescence methods that are popular in hospitals for detecting AFP. Notably, the SGGT immunosensor is also recyclable, so it has excellent potential for use in high-throughput detection.
Collapse
Affiliation(s)
- Rong Zou
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, Hubei, China
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Lei Cao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, Hubei, China
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Nan Wu
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Gang Chang
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, College of Materials Science and Engineering, Hubei University, Wuhan, 430062, China
| | - Li Li
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Lu Xiao
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Huiling Yan
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Hongjie Li
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Ping Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| | - Ting Bao
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Xiuhua Zhang
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Shengfu Wang
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China
| | - Yaping Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, Hubei, China.
| | - Hanping He
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, Hubei, China.
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China.
- College of Health Science and Engineering, Hubei University, Wuhan, 430062, Hubei, China.
| |
Collapse
|
32
|
Moreddu R. Nanotechnology and Cancer Bioelectricity: Bridging the Gap Between Biology and Translational Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304110. [PMID: 37984883 PMCID: PMC10767462 DOI: 10.1002/advs.202304110] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/25/2023] [Indexed: 11/22/2023]
Abstract
Bioelectricity is the electrical activity that occurs within living cells and tissues. This activity is critical for regulating homeostatic cellular function and communication, and disruptions of the same can lead to a variety of conditions, including cancer. Cancer cells are known to exhibit abnormal electrical properties compared to their healthy counterparts, and this has driven researchers to investigate the potential of harnessing bioelectricity as a tool in cancer diagnosis, prognosis, and treatment. In parallel, bioelectricity represents one of the means to gain fundamental insights on how electrical signals and charges play a role in cancer insurgence, growth, and progression. This review provides a comprehensive analysis of the literature in this field, addressing the fundamentals of bioelectricity in single cancer cells, cancer cell cohorts, and cancerous tissues. The emerging role of bioelectricity in cancer proliferation and metastasis is introduced. Based on the acknowledgement that this biological information is still hard to access due to the existing gap between biological findings and translational medicine, the latest advancements in the field of nanotechnologies for cellular electrophysiology are examined, as well as the most recent developments in micro- and nano-devices for cancer diagnostics and therapy targeting bioelectricity.
Collapse
|
33
|
Chang K, Fang Y, He P, Zhu C, Liu X, Zheng D, Chen D, Liu C. Employing the Anchor DSPE-PEG as a Redox Probe for Ratiometric Electrochemical Detection of Surface Proteins on Extracellular Vesicles with Aptamers. Anal Chem 2023; 95:16194-16200. [PMID: 37889159 DOI: 10.1021/acs.analchem.3c02948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Quantitative analysis of surface proteins on extracellular vesicles (EVs) has been considered to be a crucial approach for reflecting the status of diseases. Due to the diverse composition of surface proteins on EVs and the interference from nonvesicular proteins, accurately detecting the expression of surface proteins on EVs remains a challenging task. While membrane affinity molecules have been widely employed as EVs capture probes to address this issue, their inherent biochemical properties have not been effectively harnessed. In this paper, we found that the electrochemical redox activity of the DSPE-PEG molecule was diminished upon its insertion into the membrane of EVs. This observation establishes the DSPE-PEG molecule modified on the Au electrode surface as a capture and a redox probe for the electrochemical detection of EVs. By utilizing methylene blue-labeled aptamers, the targeted surface proteins of EVs can be detected by recording the ratio of the oxidation peak current of methylene blue and DSPE-PEG. Without complicated signal amplification, the detection limit for EVs is calculated to be 8.11 × 102 particles/mL. Using this platform, we directly analyzed the expression of CD63 and HER2 proteins on the surface of EVs in human clinical plasma samples, demonstrating its significant potential in distinguishing breast cancer patients from healthy individuals.
Collapse
Affiliation(s)
- Kaili Chang
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Yi Fang
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Ping He
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Chunnan Zhu
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Xiaojun Liu
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Dongyun Zheng
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Dongjuan Chen
- Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| | - Chao Liu
- The Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| |
Collapse
|
34
|
Jiang L, Lin X, Chen F, Qin X, Yan Y, Ren L, Yu H, Chang L, Wang Y. Current research status of tumor cell biomarker detection. MICROSYSTEMS & NANOENGINEERING 2023; 9:123. [PMID: 37811123 PMCID: PMC10556054 DOI: 10.1038/s41378-023-00581-5] [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: 03/10/2023] [Revised: 06/26/2023] [Accepted: 07/23/2023] [Indexed: 10/10/2023]
Abstract
With the annual increases in the morbidity and mortality rates of tumors, the use of biomarkers for early diagnosis and real-time monitoring of tumor cells is of great importance. Biomarkers used for tumor cell detection in body fluids include circulating tumor cells, nucleic acids, protein markers, and extracellular vesicles. Among them, circulating tumor cells, circulating tumor DNA, and exosomes have high potential for the prediction, diagnosis, and prognosis of tumor diseases due to the large amount of valuable information on tumor characteristics and evolution; in addition, in situ monitoring of telomerase and miRNA in living cells has been the topic of extensive research to understand tumor development in real time. Various techniques, such as enzyme-linked immunosorbent assays, immunoblotting, and mass spectrometry, have been widely used for the detection of these markers. Among them, the detection of tumor cell markers in body fluids based on electrochemical biosensors and fluorescence signal analysis is highly preferred because of its high sensitivity, rapid detection and portable operation. Herein, we summarize recent research progress in the detection of tumor cell biomarkers in body fluids using electrochemical and fluorescence biosensors, outline the current research status of in situ fluorescence monitoring and the analysis of tumor markers in living cells, and discuss the technical challenges for their practical clinical application to provide a reference for the development of new tumor marker detection methods.
Collapse
Affiliation(s)
- Liying Jiang
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
- Academy for Quantum Science and Technology, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Xinyi Lin
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Fenghua Chen
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Xiaoyun Qin
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Yanxia Yan
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Linjiao Ren
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Hongyu Yu
- Department of Mechanical and Aerospace Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Lingqian Chang
- key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083 China
| | - Yang Wang
- key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083 China
- School of Engineering Medicine, Beihang University, Beijing, 100083 China
| |
Collapse
|
35
|
Sonbhadra S, Mehak, Pandey LM. Biogenesis, Isolation, and Detection of Exosomes and Their Potential in Therapeutics and Diagnostics. BIOSENSORS 2023; 13:802. [PMID: 37622888 PMCID: PMC10452587 DOI: 10.3390/bios13080802] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/26/2023]
Abstract
The increasing research and rapid developments in the field of exosomes provide insights into their role and significance in human health. Exosomes derived from various sources, such as mesenchymal stem cells, cardiac cells, and tumor cells, to name a few, can be potential therapeutic agents for the treatment of diseases and could also serve as biomarkers for the early detection of diseases. Cellular components of exosomes, several proteins, lipids, and miRNAs hold promise as novel biomarkers for the detection of various diseases. The structure of exosomes enables them as drug delivery vehicles. Since exosomes exhibit potential therapeutic applications, their efficient isolation from complex biological/clinical samples and precise real-time analysis becomes significant. With the advent of microfluidics, nano-biosensors are being designed to capture exosomes efficiently and rapidly. Herein, we have summarized the history, biogenesis, characteristics, functions, and applications of exosomes, along with the isolation, detection, and quantification techniques. The implications of surface modifications to enhance specificity have been outlined. The review also sheds light on the engineered nanoplatforms being developed for exosome detection and capture.
Collapse
Affiliation(s)
| | | | - Lalit M. Pandey
- Bio-Interface & Environmental Engineering Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India; (S.S.); (M.)
| |
Collapse
|
36
|
Xia L, Zhang M, Hu Y, Mei W, Long Y, Wang H, Zou L, Wang Q, Yang X, Wang K. "One suction and one extrusion" mode-based wash-free platform for determination of breast cancer cell-derived exosomes. Mikrochim Acta 2023; 190:322. [PMID: 37491600 DOI: 10.1007/s00604-023-05898-6] [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: 04/05/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
A simple and wash-free POCT platform based on microcapillary was developed, using breast cancer cell-derived exosomes as a model. This method adopted the "one suction and one extrusion" mode. The hybridized complex of epithelial cell adhesion molecule (EpCAM) aptamer and complementary DNA-horseradish peroxidase conjugate (CDNA-HRP) was pre-modified on the microcapillary's inner surface. "One suction" meant inhaling the sample into the functionalized microcapillary. The exosomes could specifically bind with the EpCAM aptamer on the microcapillary's inner wall, and then the CDNA-HRP complex was released. "One extrusion" referred to squeezing the shedding CDNA-HRP into the 3,3',5,5'-tetramethylbenzidine (TMB)/H2O2 solution, and then the enzyme-catalyzed reaction would occur to make the solution yellow using sulfuric acid as the terminator. Therefore, exosome detection could be realized. The limit of detection was 2.69 × 104 particles mL-1 and the signal value had excellent linearity in the concentration range from 2.75 × 104 to 2.75 × 108 particles⋅mL-1 exosomes. In addition, the wash-free POCT platform also displayed a favorable reproducibility (RSD = 2.9%) in exosome detection. This method could effectively differentiate breast cancer patients from healthy donors. This work provided an easy-to-operate method for detecting cancer-derived exosomes without complex cleaning steps, which is expected to be applied to breast cancer screening.
Collapse
Affiliation(s)
- Ling Xia
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China
| | - Mingwan Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China
| | - Yingyun Hu
- Department of Cancer Prevention and Control, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenjing Mei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China
| | - Ying Long
- Translational Medicine Centre, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongqiang Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China
| | - Liyuan Zou
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China
| | - Qing Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China.
| | - Xiaohai Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China.
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, People's Republic of China
| |
Collapse
|
37
|
Peng W, Sun D, Lu W, Yin S, Ye B, Wang X, Ren Y, Hong Z, Zhu W, Yu P, Xi JJ, Yao B. Comprehensive Detection of PD-L1 Protein and mRNA in Tumor Cells and Extracellular Vesicles through a Real-Time qPCR Assay. Anal Chem 2023; 95:10625-10633. [PMID: 37424077 DOI: 10.1021/acs.analchem.3c00975] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
A growing number of studies have shown that tumor cells secrete extracellular vesicles (EVs) containing programmed death-ligand 1 (PD-L1) protein. These vesicles can travel to lymph nodes and remotely inactivate T cells, thereby evading immune system attack. Therefore, the simultaneous detection of PD-L1 protein expression in cells and EVs is of great significance in guiding immunotherapy. Herein, we developed a method based on qPCR for the simultaneous detection of PD-L1 protein and mRNA in EVs and their parental cells (PREC-qPCR assay). Lipid probes immobilized on magnetic beads were used to capture EVs directly from samples. For RNA assay, EVs were directly broken by heating and quantified with qPCR. As to protein assay, EVs were recognized and bound with specific probes (such as aptamers), which were used as templates in subsequent qPCR analysis. This method was used to analyze EVs of patient-derived tumor clusters (PTCs) and plasma samples from patients and healthy volunteers. The results revealed that the expression of exosomal PD-L1 in PTCs was correlated with tumor types and significantly higher in plasma-derived EVs from tumor patients than that of healthy individuals. When extended to cells and PD-L1 mRNAs, the results showed that the expression of PD-L1 protein was consistent with mRNA in cancer cell lines, while PTCs demonstrated significant heterogeneity. This comprehensive detection of PD-L1 at four levels (cell, EVs, protein, and mRNA) is believed to enhance our understanding of the relationship among PD-L1, tumors, and the immune system and to provide a promising tool for predicting the benefits of immunotherapy.
Collapse
Affiliation(s)
- Wenbo Peng
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Danyang Sun
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Wei Lu
- GeneX (Zhejiang) Precision Medicine Co., Ltd, Hangzhou 311100, China
| | - Shenyi Yin
- College of Future Technology, Peking University, Beijing 100871, China
| | - Buqing Ye
- College of Future Technology, Peking University, Beijing 100871, China
| | - Xiaoqi Wang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Yongan Ren
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Zichen Hong
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Wenyu Zhu
- Department of Oncology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Pengfei Yu
- Department of Gastric Surgery, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jianzhong Jeff Xi
- College of Future Technology, Peking University, Beijing 100871, China
| | - Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
38
|
Liu L, Tibbs J, Li N, Bacon A, Shepherd S, Lee H, Chauhan N, Demirci U, Wang X, Cunningham BT. A photonic resonator interferometric scattering microscope for label-free detection of nanometer-scale objects with digital precision in point-of-use environments. Biosens Bioelectron 2023; 228:115197. [PMID: 36905862 PMCID: PMC10072782 DOI: 10.1016/j.bios.2023.115197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/27/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023]
Abstract
Label-free detection and digital counting of nanometer-scaled objects such as nanoparticles, viruses, extracellular vesicles, and protein molecules enable a wide range of applications in cancer diagnostics, pathogen detection, and life science research. Here, we report the design, implementation, and characterization of a compact Photonic Resonator Interferometric Scattering Microscope (PRISM) designed for point-of-use environments and applications. The contrast of interferometric scattering microscopy is amplified through a photonic crystal surface, upon which scattered light from an object combines with illumination from a monochromatic source. The use of a photonic crystal substrate for interferemetric scattering microscopy results in reduced requirements for high-intensity lasers or oil-immersion objectives, thus opening a pathway toward instruments that are more suitable for environments outside the optics laboratory. The instrument incorporates two innovative elements that facilitate operation on a desktop in ordinary laboratory environments by users that do not have optics expertise. First, because scattering microscopes are extremely sensitive to vibration, we incorporated an inexpensive but effective solution of suspending the instrument's main components from a rigid metal framework using elastic bands, resulting in an average of 28.7 dBV reduction in vibration amplitude compared to an office desk. Second, an automated focusing module based on the principle of total internal reflection maintains the stability of image contrast over time and spatial position. In this work, we characterize the system's performance by measuring the contrast from gold nanoparticles with diameters in the 10-40 nm range and by observing various biological analytes, including HIV virus, SARS-CoV-2 virus, exosome, and ferritin protein.
Collapse
Affiliation(s)
- Leyang Liu
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joseph Tibbs
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Nantao Li
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Amanda Bacon
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Skye Shepherd
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hankeun Lee
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Neha Chauhan
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Stanford School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Xing Wang
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brian T Cunningham
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, Urbana, IL, 61801, USA.
| |
Collapse
|
39
|
Lv QY, Cui HF, Song X. Aptamer-based technology for gastric cancer theranostics. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:2142-2153. [PMID: 37114324 DOI: 10.1039/d3ay00415e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Gastric cancer is one of the most common causes of cancer death worldwide. This cancer exhibits high molecular and phenotype heterogeneity. The overall survival rate for gastric cancer is very low because it is always diagnosed in the advanced stages. Therefore, early detection and treatment are of great significance. Currently, biomedical studies have tapped the potential clinical applicability of aptamer-based technology for gastric cancer diagnosis and targeted therapy. Herein, we summarize the enrichment and evolution of relevant aptamers, followed by documentation of the recent developments in aptamer-based techniques for early diagnosis and precision therapy for gastric cancers.
Collapse
Affiliation(s)
- Qi-Yan Lv
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| | - Hui-Fang Cui
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| | - Xiaojie Song
- School of Life Sciences, Zhengzhou University, 100# Science Avenue, Zhengzhou 450001, People's Republic of China.
| |
Collapse
|
40
|
Lyu A, Wang Y, Cui H. Enhanced Chemiluminescence under the Nanoconfinement of Covalent-Organic Frameworks and Its Application in Sensitive Detection of Cancer Biomarkers. Anal Chem 2023; 95:7914-7923. [PMID: 37167195 DOI: 10.1021/acs.analchem.3c00372] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Chemiluminescence (CL) with intensive emission has been pursued for decades. It is still challenging to find a new mechanism to enhance CL. In this work, confinement-enhanced CL was developed for the first time by the coembedding of N-(aminobutyl)-N-(ethylisoluminol) (ABEI) and Co2+ into gold nanoparticle-modified covalent-organic frameworks (COFs). For the consideration of improving the hydrophilicity of COFs and facilitating subsequent biological modification, gold nanoparticles were first reduced on the COF surface (Au-COF) in situ without other reducing reagents. By virtue of the abundant imine bond and π backbones, ABEI and Co2+ were embedded in Au-COF synergistically through π-π stacking and coordination. The confinement of ABEI and Co2+ into Au-COF brought an over 20-fold enhancement of CL intensity compared to that of adding them to a liquid phase, which benefitted from the three aspects of the confinement effect, including the molecular enrichment effect, the physical constraint effect, and the molecular preorganization effect. As proof of concept, a lipid-protein dual-recognition sandwich strategy based on this CL-functionalized COF was developed for the detection of breast cancer cell line-derived extracellular vesicles (EVs) with four orders of magnitude improvement in the detection limit compared to ELISA. The successful distinction of human epidermal growth factor receptor 2 (HER2)-positive patients from HER2-negative patients indicated the great application potential of the proposed bioassay in HER2-positive breast cancer diagnosis. This work proposed a novel enhancement mechanism for CL based on crystalline porous materials, which provides a new perspective for the development of CL-functionalized materials for biosensors and bioassays.
Collapse
Affiliation(s)
- Aihua Lyu
- CAS Key Laboratory of Soft Matter Chemistry, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Department of Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Yisha Wang
- CAS Key Laboratory of Soft Matter Chemistry, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Department of Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Hua Cui
- CAS Key Laboratory of Soft Matter Chemistry, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), Department of Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| |
Collapse
|
41
|
Liang H, Wang X, Li F, Xie Y, Shen J, Wang X, Huang Y, Lin S, Chen J, Zhang L, Jiang B, Xing J, Zhu J. Label-free plasmonic metasensing of PSA and exosomes in serum for rapid high-sensitivity diagnosis of early prostate cancer. Biosens Bioelectron 2023; 235:115380. [PMID: 37207584 DOI: 10.1016/j.bios.2023.115380] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/15/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
Prostate-specific antigen (PSA) test is widely used to diagnose early prostate cancer (PCa). Its low sensitivity, especially in the gray zone, usually incurs overtreatment or missed diagnosis. As an emerging tumor marker, exosomes have attracted great interest in non-invasive diagnosis of PCa. However, the quick direct detection of exosomes in serum is still a big challenge for convenient screening of early PCa due to their high-degree heterogeneity and complexity. Here we develop the label-free biosensors based on wafer-scale plasmonic metasurfaces, and establish a flexible spectral methodology of exosomes profiling, which facilitates their identification and quantification in serum. We combine the metasurfaces functionalized by anti-PSA and anti-CD63, respectively, and build a portable immunoassay system to detect serum PSA and exosomes simultaneously within 20 min. Our scheme can discriminate early PCa from benign prostatic hyperplasia with a diagnostic sensitivity of 92.3%, which is much higher that of 58.3% for conventional PSA tests. The receiver operating characteristic analysis in clinical trials demonstrates significant PCa distinguishing capability with an area under the curve up to 99.4%. Our work provides a rapid and powerful approach for precise diagnosis of early PCa, and will inspire more exosomes metasensing studies for other early cancer screening.
Collapse
Affiliation(s)
- Haotian Liang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Xuegang Wang
- Department of Urology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Fajun Li
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Yinong Xie
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Jiaqing Shen
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Xueqin Wang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Yuqian Huang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Shaowei Lin
- Department of Urology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Junjie Chen
- Analysis and Measurement Center, School of Pharmaceutical Science, Xiamen University, Xiamen, 361003, China
| | - Lijian Zhang
- Department of Urology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Bingliang Jiang
- Department of Urology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Jinchun Xing
- Department of Urology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
42
|
Zhang M, Xia L, Mei W, Zou Q, Liu H, Wang H, Zou L, Wang Q, Yang X, Wang K. One-step multiplex analysis of breast cancer exosomes using an electrochemical strategy assisted by gold nanoparticles. Anal Chim Acta 2023; 1254:341130. [PMID: 37005015 DOI: 10.1016/j.aca.2023.341130] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Exosomes, as a non-invasive biomarker, perform an important role in breast cancer screening and prognosis monitoring. However, establishing a simple, sensitive, and reliable exosome analysis technique remains challenging. Herein, a one-step multiplex analysis electrochemical aptasensor based on a multi-probe recognition strategy was constructed to analyze breast cancer exosomes. HER2-positive breast cancer cell (SK-BR-3) exosomes were selected as the model targets and three aptamers including CD63, HER2 and EpCAM aptamers were used as the capture units. Methylene blue (MB) functionalized HER2 aptamer and ferrocene (Fc) functionalized EpCAM aptamer, which were modified on gold nanoparticles (Au NPs), i.e. MB-HER2-Au NPs and Fc-EpCAM-Au NPs, were used as signal units. When the mixture of target exosomes, MB-HER2-Au NPs and Fc-EpCAM-Au NPs were added on the CD63 aptamer modified gold electrode, two Au NPs modified by MB and Fc could be specifically captured on the electrode by the recognition of three aptamers with target exosomes. Then one-step multiplex analysis of exosomes was achieved by detecting two independent electrochemical signals. This strategy can not only distinguish breast cancer exosomes from other exosomes (including normal exosomes and other tumor exosomes) but also HER2-positive breast cancer exosomes and HER2-negative breast cancer exosomes. Besides, it had high sensitivity and can detect SK-BR-3 exosomes with a concentration as low as 3.4 × 103 particles mL-1. Crucially, this method can be applicable to the examination of exosomes in complicated samples, which is anticipated to afford assistance for the screening and prognosis of breast cancer.
Collapse
|
43
|
Khaksari S, Abnous K, Hadizadeh F, Ramezani M, Taghdisi SM, Mousavi Shaegh SA. Signal amplification strategies in biosensing of extracellular vesicles (EVs). Talanta 2023; 256:124244. [PMID: 36640707 DOI: 10.1016/j.talanta.2022.124244] [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: 09/17/2022] [Revised: 12/25/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022]
Abstract
Extracellular vesicles (EVs) are membrane-enclosed vesicles secreted from mammalian cells. EVs act as multicomponent delivery vehicles to carry a wide variety of biological molecular information and participate in intercellular communications. Since elevated levels of EVs are associated with some pathological states such as inflammatory diseases and cancers, probing circulating EVs holds a great potential for early diagnostics. To this end, several detection methods have been developed in which biosensors have attracted great attentions in identification of EVs due to their simple instrumentation, versatile design and portability for point-of-care applications. The concentrations of EVs in bodily fluids are extremely low (i.e. 1-100 per μl) at early stages of a disease, which necessitates the use of signal amplification strategies for EVs detection. In this way, this review presents and discusses various amplification strategies for EVs biosensors based on detection modalities including surface plasmon resonance (SPR), calorimetry, fluorescence, electrochemical and electrochemiluminescence (ECL). In addition, microfluidic systems employed for signal amplification are reviewed and discussed in terms of their design and integration with the detection methods.
Collapse
Affiliation(s)
- Sedighe Khaksari
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Laboratory of Microfluidics and Medical Microsystems, Bu Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khalil Abnous
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Ali Mousavi Shaegh
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Clinical Research Unit, Mashhad University of Medical Sciences, Mashhad, Iran; Laboratory of Microfluidics and Medical Microsystems, Bu Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
44
|
Nie Y, Wang P, Wang S, Ma Q, Su X. Accurate Capture and Identification of Exosomes: Nanoarchitecture of the MXene Heterostructure/Engineered Lipid Layer. ACS Sens 2023; 8:1850-1857. [PMID: 37114431 DOI: 10.1021/acssensors.3c00370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Recently, exosome detection has become an important breakthrough in clinical diagnosis. However, the effective capture and accurate identification of cancer exosomes in a complex biomatrix are still a tough task. Especially, the large size and non-conductivity of exosomes are not conducive to highly sensitive electrochemical or electrochemiluminescence (ECL) detection. Therefore, we have developed a Ti3C2Tx-Bi2S3-x heterostructure/engineered lipid layer-based nanoarchitecture to overcome the limitations. The engineered lipid layer not only specifically captured and efficiently fused CD63 positive exosomes but also showed excellent antifouling property in the biological matrix. Moreover, the MUC1 aptamer-modified Ti3C2Tx-Bi2S3-x heterostructure further identified and covered the gastric cancer exosomes that have been trapped in the engineered lipid layer. In the self-luminous Faraday cage-type sensing system, the Ti3C2Tx-Bi2S3-x heterostructure with sulfur vacancies extended the outer Helmholtz plane and amplified the ECL signal. Therefore, this sensor can be used to detect tumor exosomes in ascites of cancer patients without additional purification. It provides a new pathway to detect exosomes and other large-sized vesicles with high sensitivity.
Collapse
Affiliation(s)
- Yixin Nie
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Peilin Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Shuo Wang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qiang Ma
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Xingguang Su
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
45
|
Zhuang L, You Q, Su X, Chang Z, Ge M, Mei Q, Yang L, Dong W, Li L. High-Performance Detection of Exosomes Based on Synergistic Amplification of Amino-Functionalized Fe 3O 4 Nanoparticles and Two-Dimensional MXene Nanosheets. SENSORS (BASEL, SWITZERLAND) 2023; 23:3508. [PMID: 37050576 PMCID: PMC10099274 DOI: 10.3390/s23073508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
Exosomes derived from cancer cells have been recognized as a promising biomarker for minimally invasive liquid biopsy. Herein, a novel sandwich-type biosensor was fabricated for highly sensitive detection of exosomes. Amino-functionalized Fe3O4 nanoparticles were synthesized as a sensing interface with a large surface area and rapid enrichment capacity, while two-dimensional MXene nanosheets were used as signal amplifiers with excellent electrical properties. Specifically, CD63 aptamer attached Fe3O4 nanoprobes capture the target exosomes. MXene nanosheets modified with epithelial cell adhesion molecule (EpCAM) aptamer were tethered on the electrode surface to enhance the quantification of exosomes captured with the detection of remaining protein sites. With such a design, the proposed biosensor showed a wide linear range from 102 particles μL-1 to 107 particles μL-1 for sensing 4T1 exosomes, with a low detection limit of 43 particles μL-1. In addition, this sensing platform can determine four different tumor cell types (4T1, Hela, HepG2, and A549) using surface proteins corresponding to aptamers 1 and 2 (CD63 and EpCAM) and showcases good specificity in serum samples. These preliminary results demonstrate the feasibility of establishing a sensitive, accurate, and inexpensive electrochemical sensor for detecting exosome concentrations and species. Moreover, they provide a significant reference for exosome applications in clinical settings, such as liquid biopsy and early cancer diagnosis.
Collapse
Affiliation(s)
- Linlin Zhuang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Qiannan You
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Xue Su
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Zhimin Chang
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Mingfeng Ge
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Qian Mei
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Li Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Wenfei Dong
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Li Li
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| |
Collapse
|
46
|
Liu B, Wang F, Chao J. Programmable Nanostructures Based on Framework-DNA for Applications in Biosensing. SENSORS (BASEL, SWITZERLAND) 2023; 23:3313. [PMID: 36992023 PMCID: PMC10051322 DOI: 10.3390/s23063313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 06/19/2023]
Abstract
DNA has been actively utilized as bricks to construct exquisite nanostructures due to their unparalleled programmability. Particularly, nanostructures based on framework DNA (F-DNA) with controllable size, tailorable functionality, and precise addressability hold excellent promise for molecular biology studies and versatile tools for biosensor applications. In this review, we provide an overview of the current development of F-DNA-enabled biosensors. Firstly, we summarize the design and working principle of F-DNA-based nanodevices. Then, recent advances in their use in different kinds of target sensing with effectiveness have been exhibited. Finally, we envision potential perspectives on the future opportunities and challenges of biosensing platforms.
Collapse
Affiliation(s)
- Bing Liu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Fan Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| | - Jie Chao
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
47
|
Colorimetric aptasensor based on spherical nucleic acid-induced hybridization chain reaction for sensitive detection of exosomes. Talanta 2023; 258:124453. [PMID: 36924637 DOI: 10.1016/j.talanta.2023.124453] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023]
Abstract
Exosomes are one of the most promising biomarkers for tumor diagnosis and prognosis. Therefore, the development of convenient and sensitive exosome sensing strategies is of great significance. Herein, we integrated aptamer-based spherical nucleic acids (SNAs) and hybridization chain reaction (HCR) into a colorimetric aptasensor platform and applied it to the detection of exosomes. In this design, the CD63-specific aptamer pre-immobilized on the microplate was used to capture target exosomes, while the SNAs conjugated with nucleolin-specific aptamer and trigger probe H1 were designed for amplifying signal. In the presence of target exosomes, the SNAs can be attached to the microplate by the bridge effect of exosomes, resulting in the trigger of HCR. This process is accompanied by the formation of abundant G-quadruplex/hemin DNAzyme, enabling the visual quantitative analysis of exosomes. Featured with the dual amplification of SNAs and HCR, the proposed aptasensor achieved a considerable detection limit of 50 particles/μL. The practicability of this method was further verified by testing the different clinical samples. Given the ability of the aptasensor to visually detect exosomes in scenarios lacking instruments and resources, we believe that the aptasensor can be serve as a potential on-site test for liquid biopsy.
Collapse
|
48
|
Li Y, Gui Y, Zhao M, Chen X, Li H, Tian C, Zhao H, Jiang C, Xu P, Zhang S, Ye S, Huang M. The roles of extracellular vesicles in major depressive disorder. Front Psychiatry 2023; 14:1138110. [PMID: 36970289 PMCID: PMC10033661 DOI: 10.3389/fpsyt.2023.1138110] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/01/2023] [Indexed: 03/11/2023] Open
Abstract
Major depressive disorder (MDD) is a serious mental disease characterized by depressed mood, loss of interest and suicidal ideation. Its rising prevalence has rendered MDD one of the largest contributors to the global disease burden. However, its pathophysiological mechanism is still unclear, and reliable biomarkers are lacking. Extracellular vesicles (EVs) are widely considered important mediators of intercellular communication, playing an important role in many physiological and pathological processes. Most preclinical studies focus on the related proteins and microRNAs in EVs, which can regulate energy metabolism, neurogenesis, neuro-inflammation and other pathophysiological processes in the development of MDD. The purpose of this review is to describe the current research progress of EVs in MDD and highlight their potential roles as biomarkers, therapeutic indicators and drug delivery carriers for the treatment of MDD.
Collapse
Affiliation(s)
- Ying Li
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Yan Gui
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
- Department of Psychiatry, Tongde Hospital of Zhejiang Province, Mental Health Center of Zhejiang Province, Hangzhou, China
| | - Miaomiao Zhao
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Xuanqiang Chen
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Haimei Li
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Chen Tian
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Haoyang Zhao
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Chaonan Jiang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Pengfeng Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Shiyi Zhang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
| | - Shaoyong Ye
- Henan University School of Medicine, Henan University, Kaifeng, China
| | - Manli Huang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou, China
- *Correspondence: Manli Huang,
| |
Collapse
|
49
|
Wu Q, Ding Q, Lin W, Weng Y, Feng S, Chen R, Chen C, Qiu S, Lin D. Profiling of Tumor Cell-Delivered Exosome by Surface Enhanced Raman Spectroscopy-Based Biosensor for Evaluation of Nasopharyngeal Cancer Radioresistance. Adv Healthc Mater 2023; 12:e2202482. [PMID: 36528342 DOI: 10.1002/adhm.202202482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Although the advancement of radiotherapy significantly improves the survival of nasopharyngeal cancer (NPC), radioresistance associated with recurrence and poor outcomes still remains a daunting challenge in the clinical scenario. Currently, effective biomarkers and convenient detection methods for predicting radioresistance have not been well established. Here, the surface-enhanced Raman spectroscopy combined with proteomics is used to firstly profile the characteristic spectral patterns of exosomes secreted from self-established NPC radioresistance cells, and reveals specific variations of proteins expression during radioresistance formation, including collagen alpha-2 (I) chain (COL1A2) that is associated with a favorable prognosis in NPC and is negatively associated with DNA repair scores and DNA repair-related genes via bioinformatic analysis. Furthermore, deep learning model-based diagnostic model is generated to accurately identify the exosomes from radioresistance group. This work demonstrates the promising potential of exosomes as a novel biomarker for predicting the radioresistance and develops a rapid and sensitive liquid biopsy method that will provide a personalized and precise strategy for clinical NPC treatment.
Collapse
Affiliation(s)
- Qiong Wu
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, 350001, China
- College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou, Fujian, 350001, China
| | - Qin Ding
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, 350001, China
| | - Wanzun Lin
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, 201321, China
| | - Youliang Weng
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, 350001, China
| | - Shangyuan Feng
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, 350001, China
| | - Rong Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, 350001, China
| | - Chuanben Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, 350001, China
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, 350001, China
| | - Duo Lin
- Key Laboratory of OptoElectronic Science and Technology for Medicine, Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou, Fujian, 350001, China
| |
Collapse
|
50
|
Yang H, Xu H, Wang Z, Li X, Wang P, Cao X, Xu Z, Lv D, Rong Y, Chen M, Tang B, Hu Z, Deng W, Zhu J. Analysis of miR-203a-3p/SOCS3-mediated induction of M2 macrophage polarization to promote diabetic wound healing based on epidermal stem cell-derived exosomes. Diabetes Res Clin Pract 2023; 197:110573. [PMID: 36764461 DOI: 10.1016/j.diabres.2023.110573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/16/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND The development of therapeutic strategies to improve wound healing in individual diabetic patients remains challenging. Stem cell-derived exosomes represent a promising nanomaterial, and microRNAs (miRNAs) can be isolated from them. It is important to identify the potential therapeutic role of specific miRNAs, given that miRNAs can play a therapeutic role. METHODS qPCR, flow cytometry, and western blotting were used to verify the effect of epidermal stem cell-derived exosomes (EpiSC-EXOs) on M2 macrophage polarization and SOCS3 expression. By screening key miRNAs targeting SOCS3 in EpiSC-EXOs by high-throughput sequencing, we verified the mechanism in vitro. Finally, an animal model was used to verify the effect of promoting healing. RESULTS The use of EpiSC-EXOs reduced SOCS3 expression and promoted M2 macrophage polarization. The abundant miR-203a-3p present in the EpiSC-EXOs specifically bound to SOCS3 and activated the JAK2/STAT3 signaling pathway to induce M2 macrophage polarization. Treatment of the db/db mouse wound model with miR-203a-3p agomir exerted a pro-healing effect. CONCLUSIONS Our results demonstrated that the abundant miR-203a-3p present in EpiSC-EXOs can promote M2 macrophage polarization by downregulating SOCS3 and suggested that diabetic wounds can obtain better healing effects through this mechanism.
Collapse
Affiliation(s)
- Hao Yang
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Hailin Xu
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Zhiyong Wang
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Xiaohui Li
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Peng Wang
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Xiaoling Cao
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Zhongye Xu
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Dongming Lv
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Yanchao Rong
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Miao Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Bing Tang
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China
| | - Zhicheng Hu
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| | - Jiayuan Zhu
- First Affiliated Hospital of Sun Yat-sen University, Department of Burn and Wound Repair, Guangzhou, China.
| |
Collapse
|