1
|
Li JB, Yu Q, Li H, Chen J, Tian L, Fang Z, Xu Y, Yang Y, Wang S. CaCO 3 nanoparticle-encapsulated CHA circuits for sensitive fluorescence detection of miRNA in living cells. J Colloid Interface Sci 2025; 693:137578. [PMID: 40233694 DOI: 10.1016/j.jcis.2025.137578] [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: 01/07/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
MicroRNAs (miRNAs) serve as important biomarkers for various diseases, including malignant tumors, and have broad applications in diagnosis, treatment, and prognosis. The development of real-time in situ imaging methods for monitoring miRNAs has both scientific and clinical value, and in this regard, catalytic hairpin assemblies (CHAs) can be used as precise and efficient nucleic acid circuits that facilitate hybridization without depleting targets. In this study, we developed a detection system based on CHA circuits encapsulated within CaCO3 nanoparticles, which represents a novel strategy for the detection of human pancreatic cancer. This encapsulation facilitates the pH-sensitive release of DNA probes, thereby ensuring the selective and sensitive detection of cancer-associated miRNAs. Our experimental results confirmed that the fabricated nanoparticles contributed to enhancing the stability and performance of the DNA circuits, thereby enabling precise miRNA detection and effective discrimination between cancerous and non-cancerous cells. Our findings in this study highlight the potential utility of CaCO3 nanoparticle-encapsulated CHA circuits for advancing miRNA-based cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Jia-Bei Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China; Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qing Yu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hongyi Li
- College of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 402760, China
| | - Jingqi Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Linxin Tian
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhou Fang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ye Xu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
2
|
Liu L, Ju P, Yang K, Li X, Duan K, Xie J, Liu M, Chen J, Luo R. Extended Linear Confined Zipper Cascaded Reaction for Highly Efficient Intracellular Imaging and Assisting Diagnosis of Thyroid Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500202. [PMID: 40405610 DOI: 10.1002/smll.202500202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/18/2025] [Indexed: 05/24/2025]
Abstract
Highly sensitive detection and in situ tracing analysis of small-molecule biomarkers are particularly indispensable to deciphering the pathogenesis and pathological process. Despite DNA assembly-based barcoding and amplification strategies across the breadth of molecular in situ analysis, an easy-to-design, nonenzymatic, highly efficient, background leakage-avoided, highly specific, and sensitive system is highly required yet is still in its infancy. Spatial confinement nano-assembly can increase the reaction efficiency in a localized isothermal autonomous manner. Here in this work, the DNA assembly that originally relies on random collisions between freely diffusing probes is constructed between two extended linear confined probes, by which a novel confined reaction model named as extended linear confined zipper hybridization chain reaction (ZHCR) is proposed. ZHCR can significantly improve the efficiency of probe assembly and enable stable assembly within live cells, providing precise in situ target information. ZHCR system is employed to analyze two thyroid cancer-specific miRNAs, achieving in situ tracing and serum content detection. By integrating machine learning algorithms, ZHCR demonstrates significant potential in thyroid cancer auxiliary diagnosis, establishing a versatile platform that enables both highly sensitive homogeneous detection and in situ analysis of low-abundance nucleic acid fragments.
Collapse
Affiliation(s)
- Lu Liu
- Medical Examination Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pan Ju
- Medical Examination Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ketong Yang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoming Li
- Medical Examination Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kaili Duan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jingling Xie
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Department of Clinical Laboratory, Chongqing General Hospital, Chongqing, 400016, China
| | - Min Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Junman Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Rong Luo
- Medical Examination Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
3
|
Xing H, Xing X, Chen F, Li N, Xu D, Wu R, Lv Y, Li LS. DNA nanowires-mediated high sensitive quantum dot-fluorescence-linked immunoassay for proteins analysis. Anal Chim Acta 2025; 1352:343931. [PMID: 40210286 DOI: 10.1016/j.aca.2025.343931] [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: 01/17/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Highly sensitive analysis of protein biomarkers with low concentrations is essential for biological research and medical diagnosis, where quantum dots (QDs) based fluorescence-linked immunoassay (QD-FLISA) has been given considerable attention among the quantitative detection due to its outstanding characteristics. However, the traditional QD-FLISA is usually subject to the low sensitivity owing to the limited photoluminescence (PL) intensity of QDs. In this sense, the development of novel strategy that could remarkably enhance the sensitive of traditional QD-FLISA would be highly desirable. RESULTS Herein, DNA nanowires-mediated high sensitive QD-FLISA (DNA-nano-QD-FLISA) is first designed and used for the ultrasensitive detection of proteins, where DNA-nanowires are assembled through the hybridization chain reaction (HCR) and C-reactive protein (CRP) is chosen as the model analyte. The results demonstrate that the proposed DNA-nano-QD-FLISA can achieve sensitive detection of CRP, with a limit of detection (LOD) of 0.17 ng/mL, significantly lower than the system without DNA nanowires (1.66 ng/mL). Furthermore, the CRP levels in clinical samples were analyzed, yielding an excellent agreement with the Roche immunoturbidimetric method. Additionally, the versatility of the assay were demonstrated by adapting it to detect the other clinical proteins, interleukin-6 (IL-6) and procalcitonin (PCT), achieving the LODs of 0.07 ng/mL for IL-6 and 0.07 ng/mL for PCT. Furthermore, we found that the length of DNA nanowires significantly influenced the detection performance of QD-FLISA, offering a straightforward approach to precisely adjust the detection range. SIGNIFICANCE This work presents an ultra-sensitive QD-FLISA for protein detection via the introduction of DNA-nanowires assembled through HCR. The achieved results demonstrate that the incorporation of DNA nanowires enhances the detection sensitivity and accuracy of traditional QD-FLISA in quantifying low-abundance biomarkers, which holds significant clinical importance for early disease screening and diagnosis.
Collapse
Affiliation(s)
- Huanhuan Xing
- Key Lab for Special Functional Materials of Ministry of Education, and School of Nanoscience and Materials Engineering, Henan University, Kaifeng, 475004, China
| | - Xiaojing Xing
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanyang, 473061, China.
| | - Fangfang Chen
- Key Lab for Special Functional Materials of Ministry of Education, and School of Nanoscience and Materials Engineering, Henan University, Kaifeng, 475004, China
| | - Ning Li
- Key Lab for Special Functional Materials of Ministry of Education, and School of Nanoscience and Materials Engineering, Henan University, Kaifeng, 475004, China
| | - Dangdang Xu
- Key Lab for Special Functional Materials of Ministry of Education, and School of Nanoscience and Materials Engineering, Henan University, Kaifeng, 475004, China
| | - Ruili Wu
- Key Lab for Special Functional Materials of Ministry of Education, and School of Nanoscience and Materials Engineering, Henan University, Kaifeng, 475004, China
| | - Yanbing Lv
- Key Lab for Special Functional Materials of Ministry of Education, and School of Nanoscience and Materials Engineering, Henan University, Kaifeng, 475004, China.
| | - Lin Song Li
- Key Lab for Special Functional Materials of Ministry of Education, and School of Nanoscience and Materials Engineering, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
4
|
Zhang Y, Liu L, Li CM, Huang CZ, Huang J. Highly Sensitive Detection of Let-7a in Different Tumor Cells Based on a Dual Cascade Signal Amplification Strategy. Anal Chem 2025. [PMID: 40401734 DOI: 10.1021/acs.analchem.5c00865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Let-7a is a key microRNA (miRNA) cancer biomarker closely associated with cancer diagnosis and treatment, but its detection is challenging due to its small size and low abundance. In this study, we developed a highly sensitive cascade amplification strategy by integrating catalytic hairpin assembly (CHA) and hybridization chain reaction (HCR) to achieve dual-signal amplification. Two functionalized DNA tetrahedron (TDN) probes, TDN-1 and TDN-2 were first assembled by annealing four sequences and modified with four hairpins. Upon activation by let-7a, a cascade reaction occurred through the TDN-1 and TDN-2 probes, generating DNA aggregates through an isothermal CHA-HCR process. Spatial confinement effect played a key role during the CHA-HCR, enabling highly sensitive detection of let-7a while yielding a detection limit of 8.51 pM. This TDN-CHA-HCR reaction was then successfully applied to in situ imaging of endogenous let-7a in living cells, showing great promise in the early diagnosis and treatment of cancers.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Lin Liu
- Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Chun Mei Li
- Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Cheng Zhi Huang
- Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Jingtao Huang
- Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| |
Collapse
|
5
|
Zhang J, Song C, He X, Liu J, Chao J, Wang L. DNA-mediated precise regulation of SERS hotspots for biosensing and bioimaging. Chem Soc Rev 2025. [PMID: 40376876 DOI: 10.1039/d5cs00124b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Surface-enhanced Raman scattering (SERS) is a powerful analytical technique, where the creation of "hotspots" holds the key to unlocking sensitive, reproducible and reliable performance. DNA nanostructures, known for their unique predictability and exceptional programmability, have emerged as promising tools for the controllable assembly and precise regulation of SERS hotspots. In recent years, the application of DNA nanotechnology in the regulation of SERS hotspots has emerged as a research focus, but a comprehensive summary of this field is still lacking. This review begins by elucidating the mechanisms of localized surface plasmon resonance (LSPR) coupling and SERS enhancement, providing a theoretical foundation for the design principles and assembly strategies for SERS hotspots. Following this, general approaches for assembling static SERS hotspots using DNA structures of different dimensions as linkers or templates are explored. Subsequently, we delve into dynamic regulation strategies for SERS hotspots mediated by DNA structures, focusing on structural reconfiguration driven by DNA hybridization, toehold-mediated strand displacement (TMSD), and enzyme-catalyzed DNA allostery, and then summarize recent examples of DNA-mediated hotspot regulation in biosensing and bioimaging applications. Finally, we discuss future perspectives associated with the DNA-mediated precise regulation of SERS hotspots, underscoring the imperative for enhanced scalability, uniformity, and integration to pave the way for real-world applications.
Collapse
Affiliation(s)
- Jingjing Zhang
- State Key Laboratory of Flexible Electronics (LoFE), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Chunyuan Song
- State Key Laboratory of Flexible Electronics (LoFE), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Xiyu He
- State Key Laboratory of Flexible Electronics (LoFE), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Jian Liu
- State Key Laboratory of Flexible Electronics (LoFE), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Jie Chao
- State Key Laboratory of Flexible Electronics (LoFE), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Lianhui Wang
- State Key Laboratory of Flexible Electronics (LoFE), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| |
Collapse
|
6
|
Li H, Lian S, Zhang Z, Bi W, Meng Q, Ding Q. The CRISPR/Cas13a-assisted electrochemiluminescence sensing device combined with entropy-driven and hybrid chain reaction nucleic acid amplification techniques for ultra-sensitive analysis of brain natriuretic peptide. Talanta 2025; 295:128310. [PMID: 40393243 DOI: 10.1016/j.talanta.2025.128310] [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: 03/10/2025] [Revised: 04/26/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025]
Abstract
Brain natriuretic peptide (BNP) is considered a reliable marker of heart failure disease, and its timely detection can provide important pathological information to prevent or treat heart failure. In this article, an electrochemiluminescence (ECL) sensing device based on a boron carbon nitride/gold nanoparticle (BCN/AuNPs) complex is developed to determine BNP. Prominently, the CRISPR/CAS 13a enzyme reverse cleavage mode, the entropy-driven and hammer hybridization chain reaction processes were involved in the entire detection scheme. Ultimately, with multiple reaction methods and amplification reactions of nucleic acids, this ECL sensing device is able to achieve a detection limit as low as of 0.03 pg/mL and linear range from 0.1 pg/mL to 30 ng/mL for BNP. In addition, the ECL sensing device based on BCN/AuNPs complex obtained satisfactory stability and specificity, and can also be extended to the detection of other pathological markers.
Collapse
Affiliation(s)
- Haixiang Li
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Shuo Lian
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Zhiwei Zhang
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Weiye Bi
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Qingyou Meng
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
| | - Qingwei Ding
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
7
|
Li M, Zhang R, Huang X, Jian M, Qi H, Chen X, Deng J, Chen W, Feng C, Dong Q, Su B. Orthogonal DNA self-assembly technology enables rapid and accurate analysis of circulating tumor cells in breast cancer. Anal Chim Acta 2025; 1350:343871. [PMID: 40155170 DOI: 10.1016/j.aca.2025.343871] [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: 01/01/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND As a non-invasive liquid biopsy technology, the detection of circulating tumor cells (CTCs) overcomes the limitations of traditional tissue biopsy methods, enabling continuous sample collection and long-term dynamic monitoring. However, current CTCs analysis methods typically rely on cell size to separate and identify tumor cells, which fails to effectively distinguish tumor cells from different sources. In addition, existing methods are often constrained by limited antibody species, typically detecting only 2-3 molecular phenotypes. This narrow detection scope does not fully capture the heterogeneity of CTCs at the single-cell level, thus limiting its utility in precision diagnosis and personalized treatment. To address these challenges, it is urgent to develop CTCs detection methods that can simultaneously integrate comprehensive target and cell morphology information. RESULTS Using breast cancer as a research model, we developed a computer-aided design-based hybridization chain reaction (CAD-HCR) by combining DNA encoding and antibody coupling technologies with orthogonal DNA self-assembly to achieve multiple detection and heterogeneity analysis of breast cancer mimic samples. This technology overcomes the limitation of antibody species in traditional CTCs detection and utilizes antibody-trigger strand coupling to convert target protein signals into DNA signals, thereby circumventing throughput limitation of existing detection methods. By utilizing the signal amplification effect of DNA self-assembly, this technology enhances sensitivity significantly, allowing for accurate single-cell level detection of CTCs. SIGNIFICANCE This technology provides spatial positioning and cell morphological characteristics information for CTCs analysis of breast cancer, which is expected to provide a more accurate basis for diagnosis and treatment decision-making for in-depth understanding of tumor heterogeneity and clinical applications.
Collapse
Affiliation(s)
- Min Li
- School of Medicine, Anhui University of Science & Technology, Huainan, 232001, China; Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Runchi Zhang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaozhi Huang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Meng Jian
- School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Haipeng Qi
- School of Medicine, Anhui University of Science & Technology, Huainan, 232001, China; Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaolong Chen
- Shanghai Chrona Life Science Technology Co., Ltd., Shanghai, 201306, China
| | - Jie Deng
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wen Chen
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Chang Feng
- Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| | - Qiongzhu Dong
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai, 200040, China.
| | - Bin Su
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
8
|
Wong ZW, New SY. Recent Advances in Biosensors Based on Hybridization Chain Reaction and Silver Nanoclusters. SMALL METHODS 2025; 9:e2401436. [PMID: 39757735 DOI: 10.1002/smtd.202401436] [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: 09/04/2024] [Revised: 12/22/2024] [Indexed: 01/07/2025]
Abstract
Hybridization chain reaction (HCR) and DNA-templated silver nanoclusters (AgNCs) have emerged as powerful tools in biosensing. HCR enables cascade amplification through programmable DNA interactions, while DNA-AgNCs serve as transducing units with unique fluorogenic and electrochemical properties. Integrating these components into a hybrid sensor could significantly enhance sensing capabilities across various fields. Nonetheless, limited studies and the lack of systematic guidelines for HCR-AgNCs systems have hindered research progress, despite their potential. This review aims to address this gap by providing a comprehensive overview of HCR-AgNCs biosensors, facilitating further innovation in this field. The working principles, performance factors, and complementary features are discussed. Thereafter, reported HCR-AgNCs studies are assessed, emphasizing their distinct sensing mechanisms (e.g., fluorogenic, electrochemical), applications across various fields, and challenges in adopting the hybrid sensors. Drawing from the experience developing multiple HCR-AgNCs sensors, insights and guidelines for designing and developing HCR-AgNCs systems are provided for future researchers. Finally, prospective directions in HCR-AgNCs research, including multiplex assays and integration with emerging technologies, are explored to guide future advancements. The synergistic combination of HCR and AgNCs as a hybrid biosensor holds promise for addressing pressing challenges in healthcare, environmental monitoring, and beyond, paving the way for next-generation biosensing technologies.
Collapse
Affiliation(s)
- Zheng Wei Wong
- School of Pharmacy, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor Darul Ehsan, 43500, Malaysia
| | - Siu Yee New
- School of Pharmacy, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Selangor Darul Ehsan, 43500, Malaysia
| |
Collapse
|
9
|
Xie Z, Tang Z, Zhuang X, Li X, Wang B, Wang H, Zhang Y. A dual-switch fluorescence biosensor with an entropy-driven and DNA walker cascade amplification circuit for sensitive microRNA detection. NANOSCALE 2025; 17:8153-8160. [PMID: 40047714 DOI: 10.1039/d5nr00159e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
To achieve precise diagnosis of tumor cells, designing nucleic acid amplification circuits with intelligent multi-switch responsiveness for the specific and sensitive detection of miRNA within tumor cells is an important strategy. Here, we developed a dual-switch fluorescence biosensor that integrates a two-step cascade signal amplification circuit of an entropy-driven circuit (EDC) and DNA walkers onto gold nanoparticles for highly sensitive and quantitative detection of target miRNA in tumor cells. The dual switches that trigger the fluorescence signal are miRNA and the APE1 enzyme, both of which are upregulated in tumor cells. To be specific, the target miRNA21 triggers the upstream EDC, releasing strands that serve as walkers for the downstream circuit. Under the cleavage-driven action of the APE1 enzyme, the walker strands walk along the track strands on the surface of AuNPs, releasing a strong fluorescence signal and presenting good linearity in the target miRNA concentration range of 40 pM-100 nM. This biosensor presents good specificity, strong anti-interference ability against multiple RNAs and enzymes and can effectively distinguish cancer cells from normal cell lysates. Overall, this dual-switch fluorescence biosensor provides a precise recognition and efficient amplification strategy for miRNA detection within tumors, indicating its potential for clinical applications in disease diagnosis.
Collapse
Affiliation(s)
- Zikang Xie
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Zhaolong Tang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xindie Zhuang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xinhao Li
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Baozheng Wang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Hong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Yingwei Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
10
|
Li Y, Zhao Y, Yi Z, Han S. A CRISPR/Cas13a-based and hybridization chain reaction coupled evanescent wave biosensor for SARS-CoV-2 gene detection. Analyst 2025; 150:1367-1376. [PMID: 40035496 DOI: 10.1039/d4an01584c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Timely and accurate diagnosis of RNA viruses, represented by the SARS-CoV-2, is the foundation for protecting people from health threats. Currently, direct detection of viral RNA genes remains the most accurate method. Herein, a rapid, ultrasensitive, and no heating equipment required CRISPR/Cas13a based evanescent wave fluorescence biosensing platform for quantitative detection of the SARS-CoV-2 gene is reported. The collateral effect of CRISPR/Cas13a for RNA is combined with a self-driven enzyme-free hybridization chain reaction (HCR) as a signal amplification step. When the initiator RNA strand is cleaved by Cas13a, the downstream signal amplification induced by HCR is blocked, and a multiple crRNA strategy is used to enhance the cleavage efficiency. The newly designed HCR assemblies are captured by the cDNA-modified optical fiber and generate a higher-intensity fluorescence signal induced by the evanescent field. The CRISPR/Cas13a-HCR evanescent wave fluorescence biosensing platform is capable of detection of SARS-CoV-2 with a LOD of 0.47 copies per μL and the detection time is within 35 min. The spike recovery tests in saliva and high specificity have demonstrated the potential of this method for point-of-care diagnosis. This method is also suitable for the detection of other RNA viruses, without the need to alter any design of the HCR component, and only the corresponding crRNA needs to be replaced.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Yikan Zhao
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Zhihao Yi
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| | - Shitong Han
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China
| |
Collapse
|
11
|
Guo C, Gu T, Wen SH, Dang Y, Zhou Y, Ma J, Yu S. Intramolecular enhanced entropy-driven DNA-Au nanodevice for mRNA imaging in living cells. Mikrochim Acta 2025; 192:235. [PMID: 40095194 DOI: 10.1007/s00604-025-06997-2] [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: 11/26/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025]
Abstract
An intramolecular enhanced entropy-driven DNA amplifier-tethered gold nanoparticle (DNA-Au) nanodevice has been designed for highly sensitive in situ imaging of messenger ribonucleic acid (mRNA) in living cells. The DNA amplifier is immobilized on a same AuNP and the initial fluorescence of DNA-Au nanodevice is quenched. Upon internalized into the target cancer cells, the nanodevice can be activated by endogenous TK1 mRNA, and promptly release the fluorophore via the intramolecular enhanced DNA strand displacement reaction. The decreasing distance and increasing local concentration of the probes via intramolecular reaction can significantly improve the reaction kinetics of DNA-Au nanodevice, thus achieving the highly sensitive imaging of TK1 mRNA. The excellent sensitivity and selectivity allow the DNA-Au nanodevice to accurately discriminate different cell lines and monitor the variations in intracellular TK1 mRNA expression levels via fluorescence imaging. Therefore, the proposed intramolecular enhanced entropy-driven DNA-Au nanodevice will afford a reliable approach for accurate determination of mRNA in molecular diagnostic systems.
Collapse
Affiliation(s)
- Chao Guo
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
| | - Tongnian Gu
- Sunresin New Materials Co. Ltd., Xi'an Hi-tech Industrial Development Zone, Xi'an, 710076, P.R. China
| | - Shao-Hua Wen
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
| | - Yuan Dang
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
| | - Yuanzhen Zhou
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China
- Engineering Research Center of Low-Carbon Energy Efficient Utilization, Universities of Shaanxi Province, Xi'an University of Architecture and Technology, Xi'an, 710055, China
| | - Junping Ma
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China.
| | - Sha Yu
- School of Chemistry and Chemical Engineering, Xi'an University of Architecture and Technology, Xi'an, 710055, P.R. China.
| |
Collapse
|
12
|
Fdez-Sanromán A, Bernárdez-Rodas N, Rosales E, Pazos M, González-Romero E, Sanromán MÁ. Biosensor Technologies for Water Quality: Detection of Emerging Contaminants and Pathogens. BIOSENSORS 2025; 15:189. [PMID: 40136986 PMCID: PMC11940157 DOI: 10.3390/bios15030189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
This review explores the development, technological foundations, and applications of biosensor technologies across various fields, such as medicine for disease diagnosis and monitoring, and the food industry. However, the primary focus is on their use in detecting contaminants and pathogens, as well as in environmental monitoring for water quality assessment. The review classifies different types of biosensors based on their bioreceptor and transducer, highlighting how they are specifically designed for the detection of emerging contaminants (ECs) and pathogens in water. Key innovations in this technology are critically examined, including advanced techniques such as systematic evolution of ligands by exponential enrichment (SELEX), molecularly imprinted polymers (MIPs), and self-assembled monolayers (SAMs), which enable the fabrication of sensors with improved sensitivity and selectivity. Additionally, the integration of microfluidic systems into biosensors is analyzed, demonstrating significant enhancements in performance and detection speed. Through these advancements, this work emphasizes the fundamental role of biosensors as key tools for safeguarding public health and preserving environmental integrity.
Collapse
Affiliation(s)
- Antía Fdez-Sanromán
- CINTECX, Universidade de Vigo, BIOSUV, Departamento de Ingeniería Química, 36310 Vigo, Spain; (A.F.-S.); (N.B.-R.); (E.R.); (M.P.)
| | - Nuria Bernárdez-Rodas
- CINTECX, Universidade de Vigo, BIOSUV, Departamento de Ingeniería Química, 36310 Vigo, Spain; (A.F.-S.); (N.B.-R.); (E.R.); (M.P.)
| | - Emilio Rosales
- CINTECX, Universidade de Vigo, BIOSUV, Departamento de Ingeniería Química, 36310 Vigo, Spain; (A.F.-S.); (N.B.-R.); (E.R.); (M.P.)
| | - Marta Pazos
- CINTECX, Universidade de Vigo, BIOSUV, Departamento de Ingeniería Química, 36310 Vigo, Spain; (A.F.-S.); (N.B.-R.); (E.R.); (M.P.)
| | - Elisa González-Romero
- Department of Analytical and Food Chemistry, Universidade de Vigo, Campus As Lagoas-Marcosende, 36310 Vigo, Spain;
| | - Maria Ángeles Sanromán
- CINTECX, Universidade de Vigo, BIOSUV, Departamento de Ingeniería Química, 36310 Vigo, Spain; (A.F.-S.); (N.B.-R.); (E.R.); (M.P.)
| |
Collapse
|
13
|
Liu C, Huang Z, Li Z, Dai X, Chen Q, Chen M, Xu Y, Chen X, Wang F, Chen Q, Wei J. A self-sustainable DNA amplification circuit for sensitive microRNA imaging. Anal Chim Acta 2025; 1343:343689. [PMID: 39947797 DOI: 10.1016/j.aca.2025.343689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Early detection of cancer biomarkers such as microRNA-21 (miR-21), a small RNA molecule, can facilitate earlier diagnosis and potentially lead to earlier treatment. The detection of trace miRNA in complicated cellular environments necessitates the construction of self-sustainable DNA circuitry boasting high signal gain and robust anti-interference capabilities. However, current self-sustainable DNA circuits suffer from complex designs and severe signal leakage. Therefore, it is essential to develop a highly efficient and reliable strategy to improve sensing performance and accurately detect trace biomolecules in intricate biological matrices. RESULTS We engineered a general and high-performance self-sustainable DNA amplification (SDA) circuit for reliable bioimaging inside cells. The autocatalytic SDA system was composed of the catalytic DNA assembly (CDA) and the rolling circle amplification (RCA) module. Upon input of the initiator, it stimulated the self-driven cross-invasion of the CDA and RCA amplicon, facilitating the successive replication of initiator sequences and resulting in synergistically accelerated and exponential signal amplification, as systematically investigated by various experimental studies. Due to its highly efficient amplification capability and universal applicability, the self-driven SDA system enabled reliable determination of miR-21 in buffer and serum and achieved a low detection limit of 8.9 pM. As a powerful imaging strategy, the SDA circuit realized accurate miR-21 imaging within cells, highlighting its potential for clinical diagnosis. SIGNIFICANCE Reciprocal reinforcement of the CDA and RCA amplifiers accelerates the entire reaction process, facilitating the generation of an exponentially amplified FRET signal for reliable detection of analytes. The proposed SDA strategy achieves the one-step determination of DNA or miRNA with simple design, high signal gain, low signal leakage, and single-base specificity, and furthermore enables reliable miRNA localization inside cells, highlighting its potential to monitor significant biomolecules and substantially expanding the toolkits for clinical diagnosis.
Collapse
Affiliation(s)
- Chuanyi Liu
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Ziling Huang
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Zhigang Li
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Xiaojiao Dai
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Qingmin Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Min Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Yi Xu
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Xiaomei Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, 430072, Wuhan, PR China
| | - Quansheng Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China.
| | - Jie Wei
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China; College of Chemistry and Molecular Sciences, Wuhan University, 430072, Wuhan, PR China.
| |
Collapse
|
14
|
Zhang D, Zhang N, Zhao J, Li X, Bian F, Zhang Y, Ge Y, Li Z. Label-free multiplexed detection based on core-shell photonic barcodes integrated RCA. Biosens Bioelectron 2025; 271:117037. [PMID: 39657551 DOI: 10.1016/j.bios.2024.117037] [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: 08/07/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
Multiplexed, rapid, and accurate virus quantification is of great value in biomedical detection. Herein, we proposed a label-free multiplexed virus screening quantitative biosensor based on color core-shell hydrogel photonic crystal (PhC) barcode integrated rolling circle amplification (RCA). The composite hydrogel shell was formed by acrylic acid and polyethylene glycol diacrylate, and the core silica photonic crystal was used as a detector. In addition, by adjusting the internal periodic structure, the PhC microcarrier was able to perform various color barcodes for the detection of different targets. Based on these excellent properties of the nanocomposite barcode, the biosensor not only demonstrated the ability to rapidly and accurately detect SARS-COV-2-N, SARS-COV-2-S, and H1N1 simultaneously in one tube, but also converting the signal of target protein to nucleic acid signal based on DNA decorated antibody complex combine with the blocked primer and RCA strategy. As a result, the platform achieved highly sensitive multiplexed quantitative detection with a detection limit in the range of 0.30 pg/mL. In addition, the platform we developed was validated by clinical sample analysis with acceptable accuracy and high specificity, demonstrating the good potential applicability of the proposed detection method in clinical screening and diagnosis.
Collapse
Affiliation(s)
- Dagan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| | - Nan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Junqi Zhao
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Xueqin Li
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Feika Bian
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Yi Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| | - Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China.
| | - Zhiyang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| |
Collapse
|
15
|
Wei X, Ye M, Jia H, Zhou X, Wang Y, Li M, Xue C, Xu L, Shen Z. RCA-mediated tandem assembly of DNA molecular probes on lipid particles surface for efficient detection and imaging of intracellular miRNA. Biosens Bioelectron 2025; 271:116975. [PMID: 39616897 DOI: 10.1016/j.bios.2024.116975] [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: 10/09/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 01/06/2025]
Abstract
Aberrant microRNA (miRNA) expression is frequently implicated in various cancers, making the monitoring of intracellular miRNA levels a promising strategy for cancer diagnosis and therapy. However, detecting miRNA with high precision and sensitivity at the cellular level remains challenging due to its small size and low abundance. In this study, we attached hydrophobic cholesterol molecules to hydrophilic DNA chains to self-assemble into cholesterol-DNA micelles. The products of rolling ring amplification were linked to the surface of cholesterol-DNA, and two hairpins (H1 and H2) used for hybridization chain reaction (HCR) were simultaneously tethered to the branch, ultimately forming the assembled nanoprobe (RC-HCR) with signal amplification for detecting and imaging miRNA in living cells. This design significantly increased the concentration of HCR hairpins and also shortened their physical distance, thereby enhancing kinetics and signal amplification. Moreover, we demonstrated that the lipid particles could be assembled by simply stirring in a buffered solution, allowing the system to enter cells naturally. Using miR-21 as the model target, we found that the RC-HCR probe had a detection limit of 1 fM and a wide quantitative range (1 fM to 80 nM) at 37 °C within 0.5 h. In addition, RC-HCR exhibited high selectivity for miRNA detection and could accurately identify wild-type miR-21 from its mutants and other miRNAs. Furthermore, we showed that RC-HCR could efficiently image miR-21 in living cells. Collectively, our strategy provides a valuable nanoprobe for detecting and imaging miRNAs in live cells, highlighting a novel tool for early clinical diagnosis.
Collapse
Affiliation(s)
- Xiaoling Wei
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Muling Ye
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Haiyan Jia
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaoyu Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yujie Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Mengru Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chang Xue
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Liang Xu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Zhifa Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, Cixi Biomedical Research Institute, School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
16
|
Zhang R, Xie S, Yang X, Liu Y, He Y. Efficient SERS Substrate HOF@Au in Conjunction with DNAzyme-Mediated DNA Walker as a Signal Amplifier for the Ultrasensitive Detection of Lomefloxacin. Anal Chem 2025; 97:3180-3189. [PMID: 39893667 DOI: 10.1021/acs.analchem.4c06713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Lomefloxacin (LOM) is an efficacious antibiotic that might lead to liver damage and other adverse reactions if consumed over an extended period. Hence, this study proposed a DNAzyme-mediated DNA walker as a signal amplifier in combination with a substantial quantity of Au nanoparticle (NP)-loaded hydrogen-bonded organic frameworks (HOFs) as a surface-enhanced Raman scattering (SERS) substrate to accomplish the highly sensitive detection of LOM in food. Crucially, due to their high specific surface area and strong adsorption capacity, HOFs act as an ideal substrate for the in situ growth of a large amount of Au NPs, which exhibit a robust and uniform SERS enhancement. Once the target LOM was presented, the DNAzyme was activated to start the DNA walker and produce single DNA in abundance, which further triggered the catalytic hairpin assembly (CHA) cycle. In addition, the multi-interstitial core-shell structure of Au@Ag@4-nitrobenzenethiol (4-NTP)@Au@Hp3 produced more SERS "hot spots" and significantly amplified the local electromagnetic field, with HOF@Au synergistically strengthening the Raman signal of 4-NTP. Using this principle, this sensor achieved the ultrasensitive detection of LOM through the "signal on" strategy, with a detection limit as low as 4.62 × 10-14 mol/L. This strategy explored a meaningful innovative path for the design of a novel SERS biosensor for the sensitive and selective detection of antibiotics in food.
Collapse
Affiliation(s)
- Runzi Zhang
- School of Science, Xihua University, Chengdu 610039, P. R. China
| | - Shunbi Xie
- Key Laboratory for Resource Utilization of Heavy Metal Wastewater, Chongqing University of Arts and Sciences, Chongqing 402160, P. R. China
| | - Xiaoyu Yang
- School of Science, Xihua University, Chengdu 610039, P. R. China
| | - Yao Liu
- School of Food and Bioengineering, Xihua University, Chengdu 610039, P. R. China
| | - Yi He
- School of Science, Xihua University, Chengdu 610039, P. R. China
| |
Collapse
|
17
|
Malter KE, Dunbar TL, Westin C, Darin E, Alfaro JR, Shikuma NJ. A bacterial membrane-disrupting protein stimulates animal metamorphosis. mBio 2025; 16:e0357324. [PMID: 39727418 PMCID: PMC11796346 DOI: 10.1128/mbio.03573-24] [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: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Diverse marine animals undergo a metamorphic larval-to-juvenile transition in response to surface-bound bacteria. Although this host-microbe interaction is critical to establishing and maintaining marine animal populations, the functional activity of bacterial products and how they activate the host's metamorphosis program has not yet been defined for any animal. The marine bacterium Pseudoalteromonas luteoviolacea stimulates the metamorphosis of a tubeworm called Hydroides elegans by producing a molecular syringe called metamorphosis-associated contractile structures (MACs). MACs stimulate metamorphosis by injecting a protein effector termed metamorphosis-inducing factor 1 (Mif1) into tubeworm larvae. Here, we show that MACs bind to tubeworm cilia and form visible pores on the cilia membrane surface, which are smaller and less numerous in the absence of Mif1. In vitro, Mif1 associates with eukaryotic lipid membranes and possesses phospholipase activity. MACs can also deliver Mif1 to human cell lines and cause parallel phenotypes, including cell surface binding, membrane disruption, calcium flux, and mitogen-activated protein kinase activation. Finally, MACs can also stimulate metamorphosis by delivering two unrelated membrane-disrupting proteins, MLKL and RegIIIɑ. Our findings demonstrate that membrane disruption by MACs and Mif1 is necessary for Hydroides metamorphosis, connecting the activity of a bacterial protein effector to the developmental transition of a marine animal. IMPORTANCE This research describes a mechanism wherein a bacterium prompts the metamorphic development of an animal from larva to juvenile form by injecting a protein that disrupts membranes in the larval cilia. Specifically, results show that a bacterial contractile injection system and the protein effector it injects form pores in larval cilia, influencing critical signaling pathways like mitogen-activated protein kinase and calcium flux, ultimately driving animal metamorphosis. This discovery sheds light on how a bacterial protein effector exerts its activity through membrane disruption, a phenomenon observed in various bacterial toxins affecting cellular functions, and elicits a developmental response. This work reveals a potential strategy used by marine organisms to respond to microbial cues, which could inform efforts in coral reef restoration and biofouling prevention. The study's insights into metamorphosis-associated contractile structures' delivery of protein effectors to specific anatomical locations highlight prospects for future biomedical and environmental applications.
Collapse
Affiliation(s)
- Kyle E. Malter
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Tiffany L. Dunbar
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Carl Westin
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Emily Darin
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Josefa Rivera Alfaro
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| | - Nicholas J. Shikuma
- Department of Biology, San Diego State University, San Diego, California, USA
- Viral Information Institute, San Diego State University, San Diego, California, USA
| |
Collapse
|
18
|
Jiang W, Zhu Y, Tang Q, Kang X, Ji H, Guo C, Gu X, Zhang J, Mao Z, Wu L, Qin Y. On-site detection of OTA and AFB1 based on branched hybridization chain reaction coupled with lateral flow assay. Talanta 2025; 283:127095. [PMID: 39454347 DOI: 10.1016/j.talanta.2024.127095] [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: 06/27/2024] [Revised: 09/24/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Mycotoxins are widely prevalent in various agricultural commodities, whose excessive consumption can pose significant risks to human health. In this study, we developed a facile mycotoxin detection platform based on branched hybridization chain reaction coupled with lateral flow assay. Ochratoxin A/Aflatoxin B1 bind to aptamers triggering the release of initiators, which leads to bHCR amplification and forms three-dimensional dendritic DNA nanostructures. Using the functionalized quantum dots as a fluorescent label, by leveraging smartphones and handheld ultraviolet lamps, the qualitative and quantitative detection of OTA and AFB1 can be achieved with a significantly enhanced sensitivity level, surpassing that of commercial test strips by 2-3 orders of magnitude. The visual detection limits for OTA and AFB1 were 30 pg/mL and 4 pg/mL, respectively. This approach eliminates the necessity for enzyme catalysis or the preparation and purification of antibodies and/or hapten, thereby reducing testing expenses and streamlining operational procedures. Moreover, substituting aptamer and nucleic acid sequences can effectively expand the scope of detection targets. Consequently, the as-proposed strategy exhibits great potential as a versatile technique, suitable for various analytical scenarios due to its sensitivity, accuracy, simplicity, and portability.
Collapse
Affiliation(s)
- Wenjun Jiang
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Yidan Zhu
- Medical School, Nantong University, Nantong, Jiangsu, 226001, China
| | - Qu Tang
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Xiaoxia Kang
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Haiwei Ji
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China.
| | - Conglin Guo
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Xijuan Gu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Jing Zhang
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Zhenzhen Mao
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Li Wu
- School of Life Sciences, Nantong University, Nantong, Jiangsu, 226019, China.
| | - Yuling Qin
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China.
| |
Collapse
|
19
|
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
|
20
|
Wang C, Xu X, Yao W, Wang L, Pang X, Xu S, Luo X. Programmable DNA Nanoswitch-Regulated Plasmonic CRISPR/Cas12a-Gold Nanostars Reporter Platform for Nucleic Acid and Non-Nucleic Acid Biomarker Analysis Assisted by a Spatial Confinement Effect. NANO LETTERS 2025; 25:1666-1672. [PMID: 39812375 DOI: 10.1021/acs.nanolett.4c05829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
CRISPR/Cas 12a system based nucleic acid and non-nucleic acid targets detection faces two challenges including (1) multiple crRNAs are needed for multiple biomarkers detection and (2) insufficient sensitivity resulted from photobleaching of fluorescent dyes and the low kinetic cleavage rate for a traditional single-strand (ssDNA) reporter. To address these limitations, we developed a programmable DNA nanoswitch (NS)-regulated plasmonic CRISPR/Cas12a-gold nanostars (Au NSTs) reporter platform for detection of nucleic acid and non-nucleic acid biomarkers with the assistance of the spatial confinement effect. Through simply programming the target recognition sequence in NS, only one crRNA is required to detect both nucleic acid and non-nucleic acid biomarkers. The detection limit decreased by ∼196-fold for miRNA-375 and 122-fold for prostate-specific antigen (PSA), respectively. Moreover, versatile evaluation of miRNA-375 and PSA in clinical urine samples can also be achieved, according to which prostate cancer and healthy groups can be well identified.
Collapse
Affiliation(s)
- Congkai Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Xiaohan Xu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Wang Yao
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Lei Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Xiaozhe Pang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Shenghao Xu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| |
Collapse
|
21
|
Yue S, Xu X, Jiang LP, Yao H, Zhu JJ. All-In-One Entropy-Driven DNA Nanomachine for Tumor Cell-Selective Fluorescence/SERS Dual-Mode Imaging of MicroRNA. Anal Chem 2025; 97:1739-1747. [PMID: 39806536 DOI: 10.1021/acs.analchem.4c05256] [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: 01/16/2025]
Abstract
An entropy-driven catalysis (EDC) strategy is appealing for amplified bioimaging of microRNAs in living cells; yet, complex operation procedures, lacking of cell selectivity, and insufficient accuracy hamper its further applications. Here, we introduce an ingenious all-in-one entropy-driven DNA nanomachine (termed as AIO-EDN), which can be triggered by endogenous apurinic/apyrimidinic endonuclease 1 (APE1) to achieve tumor cell-selective dual-mode imaging of microRNA. Compared with the traditional EDC strategy, the integrated design of AIO-EDN achieves autocatalytic signal amplification without extra fuel strands. Moreover, the AIO-EDN leverages an endogenous APE1 overexpressed in cancer cells to activate the EDC reaction, which, however, exerts no target sensing activity in normal cells. Combining fluorescence- and surface-enhanced Raman scattering (FL/SERS) dual-mode imaging techniques, this DNA nanomachine exhibits significantly improved accuracy and tumor cell selectivity for microRNA imaging in living cells. This study provides a new paradigm to develop an integrated EDC-based platform and shows great potential in in-depth cancer diagnosis with high precision.
Collapse
Affiliation(s)
- Shuzhen Yue
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi 276005, PR China
| | - Xuan Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Li-Ping Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Huiqin Yao
- Department of Medical Chemistry, College of Basic Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| |
Collapse
|
22
|
Ke L, Dai C, Xu Y, Zhou Y. Wavelength-Resolved Magnetic Multiplex Biosensor for Simultaneous and Ultrasensitive Detection of Pneumonia Pathogens via Catalytic Hairpin Assembly Strategy with Luminescent Iridium Complexes. Anal Chem 2025; 97:1437-1445. [PMID: 39787279 DOI: 10.1021/acs.analchem.4c06318] [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: 01/12/2025]
Abstract
Pneumonia is a prevalent acute respiratory infection and a major cause of mortality and hospitalization, and the urgent demand for a rapid, direct, and highly accurate diagnostic method capable of detecting both Streptococcus pneumoniae (S. pneumoniae) and Klebsiella pneumoniae (K. pneumoniae) arises from their prominent roles as the primary pathogens responsible for pneumonia. Herein, two luminescent iridium complexes with nonoverlapping photoluminescence spectra, iridium(III)-bis [4,6-(difluorophenyl)-pyridinato-N,C2'] picolinate (abbreviated as Ir-B) and bis (2-(3,5- dimethylphenyl) quinoline-C2,N') (acetylacetonato) iridium(III)) (abbreviated as Ir-R), were unprecedently proposed to construct a novel wavelength-resolved magnetic multiplex biosensor for simultaneous detection of S. pneumoniae and K. pneumoniae based on catalytic hairpin assembly (CHA) signal amplification strategy combined with dye-doped silica nanoparticles. Notably, the proposed wavelength-resolved multiplex biosensor not only exhibits a broad linear range from 50 pM to 10 nM but also demonstrates excellent recovery rates for S. pneumoniae (96.1-99.3%) and K. pneumoniae (94.8-101.5%) in real clinical samples, with corresponding relative standard deviation (RSD) values ranging from 2.57 to 3.15% for S. pneumoniae and 1.45 to 3.17% for K. pneumoniae. These favorable experimental outcomes undoubtedly offer a promising approach for the simultaneous detection of multiple pathogens in the future.
Collapse
Affiliation(s)
- Libing Ke
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Chenji Dai
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Yaoyao Xu
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| | - Yuyang Zhou
- School of Chemistry and Life Sciences, Jiangsu Key Laboratory for Environmental Functional Materials, Suzhou University of Science and Technology, Suzhou, Jiangsu 215009, China
| |
Collapse
|
23
|
Han Y, Han S, Ren T, Han L, Ma X, Huang L, Sun X. A Dual-Cycle Isothermal Amplification Method for microRNA Detection: Combination of a Duplex-Specific Nuclease Enzyme-Driven DNA Walker with Improved Catalytic Hairpin Assembly. Int J Mol Sci 2025; 26:689. [PMID: 39859402 PMCID: PMC11766441 DOI: 10.3390/ijms26020689] [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: 12/16/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
The association between microRNAs and various diseases, especially cancer, has been established in recent years, indicating that miRNAs can potentially serve as biomarkers for these diseases. Determining miRNA concentrations in biological samples is crucial for disease diagnosis. Nevertheless, the stem-loop reverse transcription quantitative PCR method, the gold standard for detecting miRNA, has great challenges in terms of high costs and enzyme limitations when applied to clinical biological samples. In this study, an isothermal signal amplification method based on a duplex-specific nuclease (DSN) enzyme-driven DNA walker and an improved catalytic hairpin assembly (CHA) was designed for miRNA detection. First, biotin-triethylene glycol-modified trigger-releasable DNA probes were conjugated to the streptavidin-coated magnetic beads for recognizing the target miRNA. The DSN enzyme specifically hydrolyzes DNA strands when the DNA probe hybridizes with the targeted miRNA. This recycling process converts the input miRNA into short trigger fragments (catalysts). Finally, three hairpins of improved CHA are driven by this catalyst, resulting in the three-armed CHA products and a fluorescence signal as the output. This dual-cycle biosensor shows a good linear relationship in the detection of miR-21 and miR-141 over the final concentration range of 250 fM to 50 nM, presenting an excellent limit of detection (2.95 amol). This system was used to detect miR-21 and miR-141 in MCF-7 and 22RV1 cells, as well as in 1% human serum. This system can be used to evaluate the expression levels of miRNAs in different biological matrices for the clinical diagnosis and prognosis of different cancers.
Collapse
Affiliation(s)
- Yu Han
- School of Pharmaceutical Sciences, Jilin Medical University, Jilin 132013, China
| | | | | | | | | | | | - Xin Sun
- School of Pharmaceutical Sciences, Jilin Medical University, Jilin 132013, China
| |
Collapse
|
24
|
Zhan J, Li X, Feng Z, Liu Z, Feng Z, Zhu JJ, Zhang J. MicroRNA-Triggered Programmable DNA-Encoded Pre-PROTACs for Cell-Selective and Controlled Protein Degradation. Angew Chem Int Ed Engl 2025; 64:e202415323. [PMID: 39381917 DOI: 10.1002/anie.202415323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/10/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) have accelerated drug development; however, some challenges still exist owing to their lack of tumor selectivity and on-demand protein degradation. Here, we developed a miRNA-initiated assembled pre-PROTAC (miRiaTAC) platform that enables the on-demand activation and termination of target degradation in a cell type-specific manner. Using miRNA-21 as a model, we engineered DNA hairpins labeled with JQ-1 and pomalidomide and facilitated the modular assembly of DNA-encoded pre-PROTACs through a hybridization chain reaction. This configuration promoted the selective polyubiquitination and degradation of BRD4 upon miR-21 initiation, highlighting significant tumor selectivity and minimal systemic toxicity. Furthermore, the platform incorporates photolabile groups, enabling the precise optical control of pre-PROTACs during DNA assembly/disassembly, mitigating the risk of excessive protein degradation. Additionally, by introducing a secondary ligand targeting CDK6, these pre-PROTACs were used as a modular scaffold for the programmable assembly of active miRiaTACs containing two different warheads in exact stoichiometry, enabling orthogonal multitarget degradation. The integration of near-infrared light-mediated photodynamic therapy through an upconversion nanosystem further enhanced the efficacy of the platform with potent in vivo anticancer activity. We anticipate that miRiaTAC represents a significant intersection between dynamic DNA nanotechnology and PROTAC, potentially expanding the versatility of PROTAC toolkit for cancer therapy.
Collapse
Affiliation(s)
- Jiayin Zhan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun, 130022, China
| | - Xiang Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Zhe Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Zheng Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Zhiyuan Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| | - Jingjing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, China
| |
Collapse
|
25
|
Shen Y, Cai R, Wu L, Han K, Yang Y, Mao D. Programmable Intelligent DNA Nanoreactors (iDNRs) for in vivo Tumor Diagnosis and Therapy. ChemMedChem 2025; 20:e202400531. [PMID: 39377119 DOI: 10.1002/cmdc.202400531] [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: 07/14/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
With the rapid advancement of DNA technology, intelligent DNA nanoreactors (iDNRs) have emerged as sophisticated tools that harness the structural versatility and programmability of DNA. Due to their structural and functional programmability, iDNRs play an important and unique role in in vivo tumor diagnosis and therapy. This review provides an overview of the structural design methods for iDNRs based on advanced DNA technology, including enzymatic reaction-mediated and enzyme-free strategies. This review also focuses on how iDNRs achieve intelligence through functional design, as well as the applications of iDNRs for in vivo tumor diagnosis and therapy. In summary, this review summarizes current advances in iDNRs technology, discusses existing challenges, and proposes future directions for expanding their applications, which are expected to provide insights into the development of the field of in vivo tumor diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Ying Shen
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, P. R. China
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Rongkai Cai
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Liang Wu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Kun Han
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, P. R. China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Dongsheng Mao
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| |
Collapse
|
26
|
Li B, Jiang H, Luo S, Zeng Z, Xu X, Li X, Zhang S, Chen Y, Ding S, Li X, Liu J, Chen W. Enzyme-accelerated catalytic DNA circuits enable rapid and one-pot detection of bacterial pathogens. Biosens Bioelectron 2025; 267:116822. [PMID: 39362139 DOI: 10.1016/j.bios.2024.116822] [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: 07/23/2024] [Revised: 08/27/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Catalytic DNA circuits, serving as signal amplification strategies, can enable simple and accurate detection of pathogenic bacteria in complex matrices but suffer from low reaction rates and depths. Herein, we design an enzyme-accelerated catalytic hairpin assembly (EACHA) in which duplex DNA products are converted into hairpin reactants to continue participating in the next circuit reaction with the assistance of RNase H. Profiting from the high recyclability of the reactants, EACHA exhibits an approximately 37.6-fold enhancement in the rate constant and a two-order-of-magnitude improvement in sensitivity compared to conventional catalytic hairpin assembly (CHA). By integrating an allosteric probe with EACHA, a one-pot method is developed for rapid and direct detection of S. enterica Enteritidis (S. Enteritidis). This method is capable of detecting 15 CFU mL-1 of S. Enteritidis within 20 min, which is superior to that of real-time PCR. By testing 60 milk samples, we demonstrate this method's high accuracy in discriminating contaminated samples, with an area under the curve (AUC) of 0.997. Moreover, this method can be employed to accurately diagnose early-stage infected mice, with an AUC of 1.00 for feces samples and 0.986 for serum samples. Therefore, this study offers a simple and feasible method for identifying pathogens in complex matrices.
Collapse
Affiliation(s)
- Baolin Li
- School of Medicine, Xi'an Jiaotong University, 710061, Xi'an, PR China; Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 646000, Luzhou, PR China
| | - Hui Jiang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 646000, Luzhou, PR China
| | - Sijian Luo
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 646000, Luzhou, PR China
| | - Zhangrui Zeng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 646000, Luzhou, PR China
| | - Xuejing Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 646000, Luzhou, PR China
| | - Xinyu Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, 400016, Chongqing, PR China
| | - Songzhi Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, 400016, Chongqing, PR China
| | - Yirong Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, 400016, Chongqing, PR China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, 400016, Chongqing, PR China
| | - Xinmin Li
- Department of Laboratory Medicine, Chongqing Hospital of Traditional Chinese Medicine, 400011, Chongqing, PR China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, 646000, Luzhou, PR China
| | - Wei Chen
- School of Medicine, Xi'an Jiaotong University, 710061, Xi'an, PR China; Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, PR China.
| |
Collapse
|
27
|
Liu X, Zhang J, Chen Z, He X, Yan C, Lv H, Chen Z, Liu Y, Wang L, Song C. Branched hybridization chain reaction and tetrahedral DNA-based trivalent aptamer powered SERS sensor for ultra-highly sensitive detection of cancer-derived exosomes. Biosens Bioelectron 2025; 267:116737. [PMID: 39243449 DOI: 10.1016/j.bios.2024.116737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Exosomes have emerged as a promising noninvasive biomarker for early cancer diagnosis due to their ability to carry specific bioinformation related to cancer cells. However, accurate detection of trace amount of cancer-derived exosomes in complex blood remains a significant challenge. Herein, an ultra-highly sensitive SERS sensor, powered by the branched hybridization chain reaction (bHCR) and tetrahedral DNA-based trivalent aptamer (triApt-TDN), has been proposed for precise detection of cancer-derived exosomes. Taking gastric cancer SGC-7901 cells-derived exosomes as a test model, the triApt-TDNs were constructed by conjugating aptamers specific to mucin 1 (MUC1) protein with tetrahedral DNAs and subsequently immobilized on the surface of silver nanorods (AgNRs) arrays to create SERS-active sensing chips capable of specifically capturing exosomes overexpressing MUC1 proteins. The bHCR was further initiated by the trigger aptamers (tgApts) bound to exosomes, and as a result the SERS tags were assembled into AuNP network structures with abundant SERS hotspots. By optimizing the sensing conditions, the SERS sensor showed good performance in ultra-highly sensitive detection of target exosomes within 60 min detection time, with a broad response ranging of 1.44 to 1.44 × 104 particles·μL-1 and an ultralow limit of detection capable of detecting a single exosome in 2 μL sample. Furthermore, the SERS sensor exhibited good uniformity, repeatability and specificity, and capability to distinguish between gastric cancer (GC) patients and healthy controls (HC) through the detection of exosomes in clinical human serums, indicating its promising clinical potential for early diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Xinyu Liu
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Jingjing Zhang
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| | - Zeyan Chen
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Xiyu He
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Chenlong Yan
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Huiming Lv
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Zhilong Chen
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Ying Liu
- Xuzhou College of Industrial Technology, Xuzhou, 221140, China.
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| | - Chunyuan Song
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| |
Collapse
|
28
|
Nesbit KT, Hargadon AC, Renaudin GD, Kraieski ND, Buckley KM, Darin E, Lee Y, Hamdoun A, Schrankel CS. Characterization of cellular and molecular immune components of the painted white sea urchin Lytechinus pictus in response to bacterial infection. Immunol Cell Biol 2025; 103:45-59. [PMID: 39438030 PMCID: PMC11688612 DOI: 10.1111/imcb.12828] [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: 01/08/2024] [Revised: 06/07/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
Sea urchins are basal deuterostomes that share key molecular components of innate immunity with vertebrates. They are a powerful model for the study of innate immune system evolution and function, especially during early development. Here we characterize the morphology and associated molecular markers of larval immune cell types in a newly developed model sea urchin, Lytechinus pictus. We then challenge larvae through infection with an established pathogenic Vibrio and characterize phenotypic and molecular responses. We contrast these to the previously described immune responses of the purple sea urchin Strongylocentrotus purpuratus. The results revealed shared cellular morphologies and homologs of known pigment cell immunocyte markers (PKS, srcr142) but a striking absence of subsets of perforin-like macpf genes in blastocoelar cell immunocytes. We also identified novel patterning of cells expressing a scavenger receptor cysteine rich (SRCR) gene in the coelomic pouches of the larva (the embryonic stem cell niche). The SRCR signal becomes further enriched in both pouches in response to bacterial infection. Collectively, these results provide a foundation for the study of immune responses in L. pictus. The characterization of the larval immune system of this rapidly developing and genetically enabled sea urchin species will facilitate more sophisticated studies of innate immunity and the crosstalk between the immune system and development.
Collapse
Affiliation(s)
| | - Alexis Cody Hargadon
- Scripps Institution of OceanographyUniversity of California San DiegoSan DiegoCAUSA
| | - Gloria D Renaudin
- Scripps Institution of OceanographyUniversity of California San DiegoSan DiegoCAUSA
| | | | | | - Emily Darin
- Department of BiologySan Diego State UniversitySan DiegoCAUSA
| | - Yoon Lee
- Scripps Institution of OceanographyUniversity of California San DiegoSan DiegoCAUSA
| | - Amro Hamdoun
- Scripps Institution of OceanographyUniversity of California San DiegoSan DiegoCAUSA
| | | |
Collapse
|
29
|
Zhang Q, Li M, Tang Y, Zhang J, Sun C, Hao Y, Cheng J, Xie X, Jia S, Lv H, Wang F, Fan C. High-Speed Sequential DNA Computing Using a Solid-State DNA Origami Register. ACS CENTRAL SCIENCE 2024; 10:2285-2293. [PMID: 39735316 PMCID: PMC11672539 DOI: 10.1021/acscentsci.4c01557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 12/31/2024]
Abstract
DNA computing leverages molecular reactions to achieve diverse information processing functions. Recently developed DNA origami registers, which could be integrated with DNA computing circuits, allow signal transmission between these circuits, enabling DNA circuits to perform complex tasks in a sequential manner, thereby enhancing the programming space and compatibility with various biomolecules of DNA computing. However, these registers support only single-write operations, and the signal transfer involves cumbersome and time-consuming register movements, limiting the speed of sequential computing. Here, we designed a solid-state DNA origami register that compresses output data from a 3D solution to a 2D surface, establishing a rewritable register suitable for solid-state storage. We developed a heterogeneous integration architecture of liquid-state circuits and solid-state registers, reducing the register-mediated signal transfer time between circuits to less than 1 h, thereby achieving fast sequential DNA computing. Furthermore, we designed a trace signal amplifier to read surface-stored signals back into solution. This compact approach not only enhances the speed of sequential DNA computing but also lays the foundation for the visual debugging and automated execution of DNA molecular algorithms.
Collapse
Affiliation(s)
- Qian Zhang
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Mingqiang Li
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Yuqing Tang
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Jinyan Zhang
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Chenyun Sun
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Yaya Hao
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Jianing Cheng
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Xiaodong Xie
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Sisi Jia
- Zhangjiang
Laboratory, Shanghai, 201210, China
| | - Hui Lv
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
- Zhangjiang
Laboratory, Shanghai, 201210, China
| | - Fei Wang
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| | - Chunhai Fan
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, National Center
for Translational Medicine, Shanghai Jiao
Tong University, Shanghai, 200240, China
| |
Collapse
|
30
|
Hong F. Programmable DNA Reactions for Advanced Fluorescence Microscopy in Bioimaging. SMALL METHODS 2024:e2401279. [PMID: 39679773 DOI: 10.1002/smtd.202401279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/14/2024] [Indexed: 12/17/2024]
Abstract
Biological organisms are composed of billions of molecules organized across various length scales. Direct visualization of these biomolecules in situ enables the retrieval of vast molecular information, including their location, species, and quantities, which is essential for understanding biological processes. The programmability of DNA interactions has made DNA-based reactions a major driving force in extending the limits of fluorescence microscopy, allowing for the study of biological complexity at different scales. This review article provides an overview of recent technological advancements in DNA-based fluorescence microscopy, highlighting how these innovations have expanded the technique's capabilities in terms of target multiplexity, signal amplification, super-resolution, and mechanical properties. These advanced DNA-based fluorescence microscopy techniques have been widely used to uncover new biological insights at the molecular level.
Collapse
Affiliation(s)
- Fan Hong
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
31
|
Lu Z, Wang X, Chen J. AI-empowered visualization of nucleic acid testing. Life Sci 2024; 359:123209. [PMID: 39488264 DOI: 10.1016/j.lfs.2024.123209] [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: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
AIMS The visualization of nucleic acid testing (NAT) results plays a critical role in diagnosing and monitoring infectious and genetic diseases. The review aims to review the current status of AI-based NAT result visualization. It systematically introduces commonly used AI-based methods and techniques for NAT, emphasizing the importance of result visualization for accessible, clear, and rapid interpretation. This highlights the importance of developing a NAT visualization platform that is user-friendly and efficient, setting a clear direction for future advancements in making nucleic acid testing more accessible and effective for everyday applications. METHOD This review explores both the commonly used NAT methods and AI-based techniques for NAT result visualization. The focus then shifts to AI-based methodologies, such as color detection and result interpretation through AI algorithms. The article presents the advantages and disadvantages of these techniques, while also comparing the performance of various NAT platforms in different experimental contexts. Furthermore, it explores the role of AI in enhancing the accuracy, speed, and user accessibility of NAT results, highlighting visualization technologies adapted from other fields of experimentation. SIGNIFICANCE This review offers valuable insights for researchers and everyday users, aiming to develop effective visualization platforms for NAT, ultimately enhancing disease diagnosis and monitoring.
Collapse
Affiliation(s)
- Zehua Lu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 10083, China
| | - Xiaogang Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 10083, China.
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 10083, China.
| |
Collapse
|
32
|
Han Y, Li J, Li M, An R, Zhang X, Cai S. A Chemiluminescence Signal Amplification Method for MicroRNA Detection: The Combination of Molecular Aptamer Beacons with Enzyme-Free Hybridization Chain Reaction. Molecules 2024; 29:5782. [PMID: 39683939 DOI: 10.3390/molecules29235782] [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: 11/03/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
The association between microRNA (miRNA) and various diseases has been established; miRNAs have the potential to be biomarkers for these diseases. Nevertheless, the challenge of correctly quantifying an miRNA arises from its low abundance and a high degree of family homology. Therefore, in the present study, we devised a chemiluminescence (CL) detection method for miRNAs, known as the hybridization chain reaction (HCR)-CL, utilizing the enzyme-free signal amplification technology of HCR. The proposed methodology obviates the need for temperature conversion and offers a straightforward procedure owing to the absence of enzymatic participation, and the lumino-H2O2-mediated CL reaction occurs at a high rate. The technique successfully detected 2.5 amol of the target analyte and 50 amol of miR-146b in a 1% concentration of human serum. In summary, the method developed in this study is characterized by its ease of operation, cost-effectiveness, remarkable analytical prowess, and ability to detect miRNA without the need for total RNA extraction from serum samples. This method is expected to be widely used for biological sample testing in clinical settings.
Collapse
Affiliation(s)
- Yu Han
- College of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Jialin Li
- College of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Man Li
- College of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Ran An
- College of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Xu Zhang
- College of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Sheng Cai
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
33
|
Thakkar H, Thakore S, Baghel M, Kosara S, Upadhyaya H, Chalan H, Gajjar D. Ultrasensitive detection of antimicrobial resistance genes using hybridization chain reaction employing carbon dots. RSC Adv 2024; 14:38827-38831. [PMID: 39659594 PMCID: PMC11629145 DOI: 10.1039/d4ra07517j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
One of the top 10 global concerns include AntiMicrobial Resistance (AMR), which warrants the need to develop materials and methods for detection of AMR genes. Here, we propose a proof-of-concept approach for selective and ultrasensitive detection of AMR gene employing fluorescent carbon dots. Waste pistachio shell derived green emissive carbon dots (PCDs) with a high quantum yield of 24 were prepared via hydrothermal carbonization process and characterised using microscopic and spectroscopic techniques. The fluorescence-based Hybridization Chain Reaction (HCR) mediated sensing studies demonstrated the ability of the PCD sensor to detect AMR gene, compared to random and single mismatch DNA with a limit of detection of 16.17 pM. This strategy of waste valorization to design fluorescent probe offer excellent cost-effective and sustainable alternative for ultra-trace level detection of DNA.
Collapse
Affiliation(s)
- Harshil Thakkar
- Department of Chemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda 390002 India
| | - Sonal Thakore
- Department of Chemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda 390002 India
| | - Manoj Baghel
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda 390002 India
| | - Sanjay Kosara
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda 390002 India
| | - Heli Upadhyaya
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda 390002 India
| | - Hemanta Chalan
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda 390002 India
| | - Devarshi Gajjar
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda 390002 India
| |
Collapse
|
34
|
Jiang Y, Chen Y, Shang J, Yu M, Weng B, Liu J, Liu X, Wang F. On-Site Multiply Stimulated Self-Confinement of an Integrated DNA Cascade Circuit for Highly Reliable Intracellular Imaging of miRNA and In Situ Interrogation of the Relevant Regulatory Pathway. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406545. [PMID: 39282814 DOI: 10.1002/smll.202406545] [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: 07/31/2024] [Revised: 08/31/2024] [Indexed: 12/06/2024]
Abstract
Artificial DNA circuits represent a versatile yet promising toolbox for in situ monitoring and concomitant regulation of diverse biological events within live cells. Nonetheless, their performance is significantly impeded by the diffusion-dominated slow reaction kinetics and the uncontrollable off-target activation. Herein, a self-localized cascade (SLC) circuit is reported for the robust and efficient microRNA (miRNA) analysis in living cells. The SLC circuit consists of the cell-specific localization module and the analyte-specific signal amplification module. By integrating the reaction probes of these two modules, the complexity of the system is reduced to realize the responsive co-localization of circuitry probes and the simultaneous cascade signal amplification. Taking advantage of the specifically activated, self-localized, and cascade design, the SLC circuit successfully achieves the robust miRNA-21 (miR-21) imaging and the accurate cells differentiation. Moreover, the reverse regulation mechanism is successfully explored between messenger RNA (mRNA) and miRNA through the engineered SLC circuit and further elucidates the underlying signaling pathways between them. Therefore, the SLC circuit provides a powerful tool for the sensitive detection of intracellular biomolecules and the study of the corresponding cell regulatory mechanisms.
Collapse
Affiliation(s)
- Yuqian Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yingying Chen
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Jinhua Shang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Mengdi Yu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Benrui Weng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Jing Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
- Department of Gastroenterology, Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
- Department of Gastroenterology, Hubei Provincial Clinical Research Center for Intestinal and Colorectal Diseases, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| |
Collapse
|
35
|
Yang Y, Zhang N, Jiang W. Functional DNA-Zn 2+ coordination nanospheres for sensitive imaging of 8-oxyguanine DNA glycosylase activity in living cells. Talanta 2024; 280:126779. [PMID: 39217713 DOI: 10.1016/j.talanta.2024.126779] [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: 07/16/2024] [Revised: 08/15/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Sensitive monitoring of human 8-oxyguanine DNA glycosylase (hOGG1) activity in living cells is helpful to understand its function in damage repair and evaluate its role in disease diagnosis. Herein, a functional DNA-Zn2+ coordination nanospheres was proposed for sensitive imaging of hOGG1 in living cells. The nanospheres were constructed through the coordination-driven self-assembly of the entropy driven reaction (EDR) -deoxyribozyme (DNAzyme) system with Zn2+, where DNAzyme was designed to split structure and assembled into the EDR system. When the nanospheres entered the cell, the competitive coordination between phosphate in the cell and Zn2+ leaded to the disintegration of the nanospheres, releasing DNA and some Zn2+. The released Zn2+ acted as a cofactor of DNAzyme. In the presence of hOGG1, the EDR was completed, accompanied by fluorescence recovery and the generation of a complete DNAzyme. With the assistance of Zn2+, DNAzyme continuously cleaved substrates to produce plenty of fluorescence signals, thus achieving sensitive imaging of hOGG1 activity. The nanospheres successfully achieved sensitive imaging of hOGG1 in human cervical cancer cells (HeLa), human non-small cell lung cancer cells and human normal colonic epithelial cells, and assayed changes in hOGG1 activity in HeLa cells. This nanospheres may provide a new tool for intracellular hOGG1 imaging and related biomedical studies.
Collapse
Affiliation(s)
- Yayun Yang
- School of Chemistry and Chemical Engineering, Shandong University, 250100, Jinan, PR China
| | - Nan Zhang
- Research Center of Basic Medicine, Breast Center, Central Hospital Affiliated to Shandong First Medical University, 250013, Jinan, PR China.
| | - Wei Jiang
- School of Chemistry and Chemical Engineering, Shandong University, 250100, Jinan, PR China.
| |
Collapse
|
36
|
Qiu J, Liu C, Zhu Y. Rolling circle amplification cooperating crRNA switch for direct and sensitive methicillin-resistant Staphylococcus aureus (MRSA) analysis. Biotechnol Lett 2024; 47:4. [PMID: 39585465 DOI: 10.1007/s10529-024-03550-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/28/2024] [Accepted: 10/19/2024] [Indexed: 11/26/2024]
Abstract
Evaluating the methicillin resistance of Staphylococcus aureus (S. aureus) is highly important for adapting nursing strategies. Nevertheless, the identification of methicillin-resistant S. aureus (MRSA) that is both sensitive and reliable continues to pose a significant obstacle. This study describes a method for detecting MRSA using a combination of fixed rolling circle amplification (RCA) and the exonuclease-iii (Exo-iii) assisted CRISPR-Cas12a system for signal amplification. When MRSA is present, the interaction between the "b" chain in the capture probe and MRSA allows the "a" chain to be exposed. This "a" chain acts as a primer to initiate the fixed RCA process. The H probe, which includes the crRNA segment, forms a bond with the RCA product and then releases the crRNA segment with the aid of Exo-iii. The Cas12a protein, when combined with the crRNA, generates an activated CRISPR-Cas12a system that cleaves the "Reporter" probe, resulting in the production of fluorescent signals. Furthermore, this fluorescent test has been utilized for the examination of clinical samples with a satisfactory rate of retrieval. Based on the elegant design, the proposed method exhibited a low detection limit of 4.6 cfu/mL, while maintaining a high specificity for MRSA even from a mixture of several interfering bacteria. Due to its cost-effectiveness, simplicity, and adaptability, the sensing system shows potential as a platform for detecting MRSA and evaluating postoperative nursing for stomach cancer patients.
Collapse
Affiliation(s)
- Junling Qiu
- Department of Gastroenterology, People's Hospital Of Chongqing Liang Jiang New Area, No. 199 Renxing Road, Renhe Street, Yubei District, Chongqing, 401147, China
| | - Chang Liu
- Department of Gastroenterology, People's Hospital Of Chongqing Liang Jiang New Area, No. 199 Renxing Road, Renhe Street, Yubei District, Chongqing, 401147, China
| | - Yuxia Zhu
- Department of Gastroenterology, People's Hospital Of Chongqing Liang Jiang New Area, No. 199 Renxing Road, Renhe Street, Yubei District, Chongqing, 401147, China.
| |
Collapse
|
37
|
Cao X, Cai J, He Z, Ji H, Sun R, Zhang X, Chen C, Zhu Q. A strategy for detecting CSFV using DNAzyme-HCR cascade amplification. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:7772-7780. [PMID: 39403851 DOI: 10.1039/d4ay01209g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
The Hybridization Chain Reaction (HCR) is an isothermal amplification technique widely used for sensing nucleic acids and small molecules. Despite its effectiveness, conventional linear HCR exhibits relatively slow kinetics and insufficient sensitivity. To address this challenge, we have innovatively combined HCR with DNAzyme technology to enhance nucleic acid detection. In this novel approach, the presence of a target molecule triggers the formation of DNAzyme, leading to the cleavage of substrate S, the initiation of HCR, and the production of DNA nanowires and labeled DNAzyme. The newly generated DNAzyme continuously cleaves substrate S, promoting sequential HCR amplification and significantly enhancing the fluorescence signal. This system offers a simple, sensitive, selective, and versatile method for nucleic acid detection, with a detection limit as low as 5 pM. When tested on classical swine fever virus (CSFV) samples, the system demonstrated detection accuracy comparable to RT-qPCR and exhibited superior repeatability.
Collapse
Affiliation(s)
- Xiuen Cao
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| | - Jiajing Cai
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| | - Zhilin He
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| | - Haofei Ji
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| | - Ruowei Sun
- Hunan Zaochen Nanorobot Co. Ltd, Liuyang 410300, Hunan, China.
| | - Xun Zhang
- Hunan Zaochen Nanorobot Co. Ltd, Liuyang 410300, Hunan, China.
| | - Chuanpin Chen
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| | - Qubo Zhu
- Xiangya School of Pharmaceutical Sciences in Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
38
|
Chu M, Zhang Y, Ji C, Zhang Y, Yuan Q, Tan J. DNA Nanomaterial-Based Electrochemical Biosensors for Clinical Diagnosis. ACS NANO 2024; 18:31713-31736. [PMID: 39509537 DOI: 10.1021/acsnano.4c11857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Sensitive and quantitative detection of chemical and biological molecules for screening, diagnosis and monitoring diseases is essential to treatment planning and response monitoring. Electrochemical biosensors are fast, sensitive, and easy to miniaturize, which has led to rapid development in clinical diagnosis. Benefiting from their excellent molecular recognition ability and high programmability, DNA nanomaterials could overcome the Debye length of electrochemical biosensors by simple molecular design and are well suited as recognition elements for electrochemical biosensors. Therefore, to enhance the sensitivity and specificity of electrochemical biosensors, significant progress has been made in recent years by optimizing the DNA nanomaterials design. Here, the establishment of electrochemical sensing strategies based on DNA nanomaterials is reviewed in detail. First, the structural design of DNA nanomaterial is examined to enhance the sensitivity of electrochemical biosensors by improving recognition and overcoming Debye length. In addition, the strategies of electrical signal transduction and signal amplification based on DNA nanomaterials are reviewed, and the applications of DNA nanomaterial-based electrochemical biosensors and integrated devices in clinical diagnosis are further summarized. Finally, the main opportunities and challenges of DNA nanomaterial-based electrochemical biosensors in detecting disease biomarkers are presented in an aim to guide the design of DNA nanomaterial-based electrochemical devices with high sensitivity and specificity.
Collapse
Affiliation(s)
- Mengge Chu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yawen Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Cailing Ji
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yun Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Quan Yuan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Jie Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
39
|
Zhao X, Wang Z, Zhang H, Liu J, Wu W, Yu L, Xu C, Wang X, Hu Q. Highly Sensitive One-Pot Isothermal Assay Combining Rolling Circle Amplification and CRISPR/Cas12a for Aflatoxin B 1 Detection. Anal Chem 2024; 96:18070-18078. [PMID: 39491486 DOI: 10.1021/acs.analchem.4c03798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Occurrences of mycotoxins in cereals are widespread throughout the world. However, the lack of efficient and ultrasensitive tests has largely impeded the identification of these substances in actual samples. Herein, a novel one-pot isothermal assay that integrates rolling-circle amplification (RCA) and CRISPR/Cas12a to detect aflatoxin B1 (AFB1) is reported. Upon addition of AFB1 to the magnetic bead functionalized with a duplex of the AFB1 aptamer and its complementary DNA (cDNA), the specific recognition of AFB1 by the aptamer causes the release of cDNA to activate the RCA reaction. Subsequently, the RCA amplicon initiates both trans-cleavage and cis-cleavage activities of the endonuclease Cas12a. The synergistic coupling of RCA and CRISPR/Cas12a enables exponential amplification of cDNA, which further promotes CRISPR/Cas12a to nonspecifically cleave the single-stranded DNA reporters with enhanced detection signals. Remarkably, the CRISPR/Cas12a-assisted one-pot isothermal assay can not only achieve ultrasensitive quantitative detection through fluorescence detection, but also achieve visual detection through a lateral flow strip, which improves accessibility to mycotoxin detection in resource-limited regions. The limit of detection was 0.016 and 0.408 ng/mL, respectively. The proposed assay successfully applies in real samples with satisfactory recoveries from 90 to 114%. This study presents a powerful and versatile method for reliable and ultrasensitive detection of mycotoxins in various applications.
Collapse
Affiliation(s)
- Xinxin Zhao
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Zhongxing Wang
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Hao Zhang
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Jinpeng Liu
- Key Laboratory of Colloid and Interface Chemistry, Shandong University, Ministry of Education, Jinan 250100, China
| | - Wenli Wu
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Li Yu
- Key Laboratory of Colloid and Interface Chemistry, Shandong University, Ministry of Education, Jinan 250100, China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiao Wang
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Qiongzheng Hu
- Qilu University of Technology (Shandong Academy of Sciences), Shandong Analysis and Test Center, Jinan 250014, China
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| |
Collapse
|
40
|
Wu Q, Xu W, Shang J, Li J, Liu X, Wang F, Li J. Autocatalytic DNA circuitries. Chem Soc Rev 2024; 53:10878-10899. [PMID: 39400237 DOI: 10.1039/d4cs00046c] [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: 10/15/2024]
Abstract
Autocatalysis, a self-sustained replication process where at least one of the products functions as a catalyst, plays a pivotal role in life's evolution, from genome duplication to the emergence of autocatalytic subnetworks in cell division and metabolism. Leveraging their programmability, controllability, and rich functionalities, DNA molecules have become a cornerstone for engineering autocatalytic circuits, driving diverse technological applications. In this tutorial review, we offer a comprehensive survey of recent advances in engineering autocatalytic DNA circuits and their practical implementations. We delve into the fundamental principles underlying the construction of these circuits, highlighting their reliance on DNAzyme biocatalysis, enzymatic catalysis, and dynamic hybridization assembly. The discussed autocatalytic DNA circuitry techniques have revolutionized ultrasensitive sensing of biologically significant molecules, encompassing genomic DNAs, RNAs, viruses, and proteins. Furthermore, the amplicons produced by these circuits serve as building blocks for higher-order DNA nanostructures, facilitating biomimetic behaviors such as high-performance intracellular bioimaging and precise algorithmic assembly. We summarize these applications and extensively address the current challenges, potential solutions, and future trajectories of autocatalytic DNA circuits. This review promises novel insights into the advancement and practical utilization of autocatalytic DNA circuits across bioanalysis, biomedicine, and biomimetics.
Collapse
Affiliation(s)
- Qiong Wu
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Wei Xu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jinhua Shang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Jiajing Li
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xiaoqing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Fuan Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Jinghong Li
- Department of Chemistry, Center for Bioanalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
- Beijing Institute of Life Science and Technology, Beijing 102206, China
| |
Collapse
|
41
|
Wang Y, Tian R, Li Z, Ma S, Wu Y, Liu F, Han Q, Li J, Zhao RC, Jiang Q, Ding B. Mesenchymal Stem Cells Engineered by Multicomponent Coassembled DNA Nanofibers for Enhanced Wound Healing. NANO LETTERS 2024; 24:13955-13964. [PMID: 39445643 DOI: 10.1021/acs.nanolett.4c03144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
A major challenge for stem cell therapies, such as using mesenchymal stem cells to treat skin injuries, is the stable engraftment of exogenous cells and the maintenance of their regenerative capacities in the wound areas. DNA-based self-assembly strategies can be used for artificial and multifunctional cell surface engineering to stabilize and enhance their functions for therapeutic applications. Here, we developed DNA nanofiber-decorated stem cells, in which DNA-based, multivalent fiber-like structures were self-assembled in situ on the cell surfaces. These engineered stem cells have demonstrated robust reactive oxygen species (ROS) scavenging effects, specific adhesion to damaged vascular endothelial cells, and the ability to enhance angiogenesis, which were effective and safe for acute or chronic wound healing in a mouse model with excisional skin injury. This DNA nanostructure-engineered stem cell provides a novel therapeutic platform for the treatment of tissue damage.
Collapse
Affiliation(s)
- Yiming Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College; Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences; Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing 100005, China
| | - Run Tian
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuoting Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College; Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences; Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing 100005, China
| | - Shuaijing Ma
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College; Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences; Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing 100005, China
| | - Yushuai Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengsong Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College; Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences; Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing 100005, China
| | - Jing Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College; Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences; Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing 100005, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College; Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences; Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing 100005, China
| | - Qiao Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
42
|
Zheng H, Li X, Liu X, Xu X. Controlling the Depth of Hybridization Chain Reaction by Extended Dangling Ends and Its Analytical Applications. Anal Chem 2024; 96:17054-17058. [PMID: 39404142 DOI: 10.1021/acs.analchem.4c03841] [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: 10/30/2024]
Abstract
Hybridization chain reaction (HCR) is a powerful enzyme-free nucleic acid amplification strategy. Triggered by an initiator strand, it yields nicked double helices analogous to alternating copolymers. However, there is no effective way to regulate the HCR reaction, and the most apparent phenomenon is the uncontrollable polymerization of product after introducing an initiator. Here we explore controlling the depth of the HCR reaction by extended dangling ends on hairpin monomers and report that sequence length, nucleotide composition, and secondary structure can alter HCR polymerization and can be utilized for the desired regulation. Interaction dynamics between initiator and hairpin monomers simulated by oxDNA are in good accordance with experimental results. Such a controlling effect can be utilized for new analytical applications that HCR cannot previously achieve, such as analyzing strand-extension enzymes and identifying short-sequence structures. The finding provides a concise but effective way for controlling the depth of HCR reaction and opens the application scope of HCR to more fields.
Collapse
Affiliation(s)
- Hongzheng Zheng
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - Xuesi Li
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - Xiuqian Liu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - Xiaowen Xu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| |
Collapse
|
43
|
Feng Y, Yang J, He Z, Liu X, Ma C. CRISPR-Cas-based biosensors for the detection of cancer biomarkers. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:6634-6653. [PMID: 39258950 DOI: 10.1039/d4ay01446d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Along with discovering cancer biomarkers, non-invasive detection methods have played a critical role in early cancer diagnosis and prognostic improvement. Some traditional detection methods have been used for detecting cancer biomarkers, but they are time-consuming and involve materials and human costs. With great flexibility, sensitivity and specificity, the clustered regularly interspaced short palindromic repeats (CRISPR)-associated system provides a wide range of application prospects in this field. Herein, we introduce the background of the CRISPR-Cas (CRISPR-associated) system and comprehensively summarize the diagnosis strategies of cancer mediated by the CRISPR-Cas system, including four kinds of biochemical-based markers: nucleic acid, enzyme, tumor-specific protein and exosome. Furthermore, we discuss the challenges in implementing the CRISPR-Cas system in clinical applications.
Collapse
Affiliation(s)
- Yuxin Feng
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jinmeng Yang
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Ziping He
- School of Life Sciences, Central South University, Changsha 410013, China.
- Clinical Medicine Eight-year Program, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Xinfa Liu
- School of Life Sciences, Central South University, Changsha 410013, China.
| | - Changbei Ma
- School of Life Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
44
|
Diao N, Hou J, Peng X, Wang Y, He A, Gao H, Yang L, Guo P, Wang J, Han D. Multiplexed and Quantitative Imaging of Live-Cell Membrane Proteins by a Precise and Controllable DNA-Encoded Amplification Reaction. Angew Chem Int Ed Engl 2024; 63:e202406330. [PMID: 38979704 DOI: 10.1002/anie.202406330] [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: 04/12/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/10/2024]
Abstract
Amplifying DNA conjugated affinity ligands can improve the sensitivity and multiplicity of cell imaging and play a crucial role in comprehensively deciphering cellular heterogeneity and dynamic changes during development and disease. However, the development of one-step, controllable, and quantitative DNA amplification methods for multiplexed imaging of live-cell membrane proteins is challenging. Here, we introduce the template adhesion reaction (TAR) method for assembling amplifiable DNA sequences with different affinity ligands, such as aptamers or antibodies, for amplified and multiplexed imaging of live-cell membrane proteins with high quantitative fidelity. The precisely controllable TAR enables proportional amplification of membrane protein targets with variable abundances by modulating the concentration ratios of hairpin templates and primers, thus allowing sensitive visualization of multiple membrane proteins with enhanced signal-to-noise ratios (SNRs) without disturbing their original ratios. Using TAR, we achieved signal-enhanced imaging of six proteins on the same live-cell within 1-2 h. TAR represents an innovative and programmable molecular toolkit for multiplexed profiling of membrane proteins in live-cells.
Collapse
Affiliation(s)
- Nannan Diao
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jianing Hou
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xinyu Peng
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- College of Life Science, Shanghai University, Shanghai, 200444, China
| | - Yaru Wang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Axin He
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Haiyan Gao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
| | - Linlin Yang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Pei Guo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Junyan Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Da Han
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
45
|
Corujo-Simon E, Bates LE, Yanagida A, Jones K, Clark S, von Meyenn F, Reik W, Nichols J. Human trophectoderm becomes multi-layered by internalization at the polar region. Dev Cell 2024; 59:2497-2505.e4. [PMID: 38889726 DOI: 10.1016/j.devcel.2024.05.028] [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: 10/11/2023] [Revised: 02/02/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024]
Abstract
To implant in the uterus, mammalian embryos form blastocysts comprising trophectoderm (TE) surrounding an inner cell mass (ICM), confined to the polar region by the expanding blastocoel. The mode of implantation varies between species. Murine embryos maintain a single layered TE until they implant in the characteristic thick deciduum, whereas human blastocysts attach via polar TE directly to the uterine wall. Using immunofluorescence (IF) of rapidly isolated ICMs, blockade of RNA and protein synthesis in whole embryos, or 3D visualization of immunostained embryos, we provide evidence of multi-layering in human polar TE before implantation. This may be required for rapid uterine invasion to secure the developing human embryo and initiate formation of the placenta. Using sequential fluorescent labeling, we demonstrate that the majority of inner TE in human blastocysts arises from existing outer cells, with no evidence of conversion from the ICM in the context of the intact embryo.
Collapse
Affiliation(s)
- Elena Corujo-Simon
- Wellcome - MRC Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK.
| | - Lawrence Edward Bates
- Wellcome - MRC Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Ayaka Yanagida
- Wellcome - MRC Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Kenneth Jones
- Wellcome - MRC Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Stephen Clark
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | | | - Wolf Reik
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | - Jennifer Nichols
- Wellcome - MRC Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 3EG, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
46
|
Fu S, Li J, Chen J, Zhang L, Liu J, Liu H, Su X. Bacteriophage λ exonuclease and a 5'-phosphorylated DNA guide allow PAM-independent targeting of double-stranded nucleic acids. Nat Biotechnol 2024:10.1038/s41587-024-02388-9. [PMID: 39294394 DOI: 10.1038/s41587-024-02388-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/19/2024] [Indexed: 09/20/2024]
Abstract
Sequence-specific recognition of double-stranded nucleic acids is essential for molecular diagnostics and in situ imaging. Clustered regularly interspaced short palindromic repeats and Cas systems rely on protospacer-adjacent motif (PAM)-dependent double-stranded DNA (dsDNA) recognition, limiting the range of targetable sequences and leading to undesired off-target effects. Using single-molecule fluorescence resonance energy transfer analysis, we discover the enzymatic activity of bacteriophage λ exonuclease (λExo). We show binding of 5'-phosphorylated single-stranded DNA (pDNA) to complementary regions on dsDNA and DNA-RNA duplexes, without the need for a PAM-like motif. Upon binding, the λExo-pDNA system catalytically digests the pDNA into nucleotides in the presence of Mg2+. This process is sensitive to mismatches within a wide range of the pDNA-binding region, resulting in exceptional sequence specificity and reduced off-target effects in various applications. The absence of a requirement for a specific motif such as a PAM sequence greatly broadens the range of targets. We demonstrate that the λExo-pDNA system is a versatile tool for molecular diagnostics, DNA computing and gene imaging applications.
Collapse
Affiliation(s)
- Shengnan Fu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering and State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Junjie Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering and State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jing Chen
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering and State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Linghao Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering and State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Jiajia Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering and State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering and State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xin Su
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering and State Key Laboratory of Organic-Inorganic Composites, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
47
|
Liu S, Wang J, Chen Y, Fan J, Du B, Liu R, Zhu X, Wang K, Xie N, Huang J. Modular Assembled Localized Hybridization Chain Reaction for In Situ mRNA Amplified Imaging. NANO LETTERS 2024; 24:11590-11598. [PMID: 39225632 DOI: 10.1021/acs.nanolett.4c03099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
As a nonenzymatic DNA signal amplification technique, localized hybridization chain reaction (LHCR) was designed to improve the limitations in response speed and low sensitivity of conventional free diffusional HCR (hybridization chain reaction). However, it is still confronted with the challenges of complicated DNA scaffolds with low loading capacity and a time-consuming process of diffusion. Herein, we introduced modular assembly of a DNA minimal scaffold for coassembly of DNA hairpins for amplified fluorescence imaging of mRNA in situ. DNA hairpins were spatially bound to two Y-shaped modules to form H-shaped DNA modules, and then multiple H-shaped DNA modules can further assemble into an H-module-based hairpin scaffold (HHS). Benefiting from highly spatial localization and high loading capacity, the HHS system showed higher sensitivity and faster speed. It has also been proven to work perfectly in vitro and in vivo, which could provide a promising bioanalysis system for low abundance biomolecule detection.
Collapse
Affiliation(s)
- Shiyuan Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Jiaoli Wang
- School of Electrical Engineering, University of South China, Hengyang 421001, P. R. China
| | - Yu Chen
- 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, P. R. China
| | - Jiahao Fan
- 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, P. R. China
| | - Bin Du
- 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, P. R. China
| | - Ruiting Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China
| | - Xiaobei Zhu
- 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, P. R. 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, P. R. China
| | - Nuli Xie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, P. R. China
| | - Jin Huang
- 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, P. R. China
| |
Collapse
|
48
|
Gu A, Dong Y, Li L, Yu D, Zhang J, Chen Y. CRISPR/Cas12a and Hybridization Chain Reaction-Coregulated Magnetic Relaxation Switching Biosensor for Sensitive Detection of Viable Salmonella in Animal-Derived Foods. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20130-20139. [PMID: 39192723 DOI: 10.1021/acs.jafc.4c05540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
We combined a CRISPR/Cas12a system with a hybridization chain reaction (HCR) to develop an ultrasensitive magnetic relaxation switching (MRS) biosensor for detecting viable Salmonella typhimurium (S. typhimurium). Magnetic nanoparticles of two sizes (30 and 1000 nm: MNP30 and MNP1000, respectively) were coupled through HCR. The S. typhimurium gene-activated CRISPR/Cas12a system released MNP30 from the MNP1000-HCR-MNP30 complex through a trans-cleavage reaction. After magnetic separation, released MNP30 was collected from the supernatant and served as a transverse relaxation time (T2) signal probe. Quantitative detection of S. typhimurium is achieved by establishing a linear relationship between the change in T2 and the target gene. The biosensor's limit of detection was 77 CFU/mL (LOD = 3S/M, S = 22.30, M = 0.87), and the linear range was 102-108 CFU/mL. The accuracy for detecting S. typhimurium in real samples is comparable to that of qPCR. Thus, this is a promising method for the rapid and effective detection of foodborne pathogens.
Collapse
Affiliation(s)
- Aoting Gu
- College of Food Science and Technology, Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Yongzhen Dong
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, Liaoning 116034, China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Letian Li
- College of Food Science and Technology, Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
| | - Deyang Yu
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, Liaoning 116034, China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jiangjiang Zhang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yiping Chen
- College of Food Science and Technology, Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei 430070, China
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, Liaoning 116034, China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| |
Collapse
|
49
|
Long D, Shi P, Xu X, Ren J, Chen Y, Guo S, Wang X, Cao X, Yang L, Tian Z. Understanding the relationship between sequences and kinetics of DNA strand displacements. Nucleic Acids Res 2024; 52:9407-9416. [PMID: 39077949 PMCID: PMC11381357 DOI: 10.1093/nar/gkae652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/18/2024] [Accepted: 07/14/2024] [Indexed: 07/31/2024] Open
Abstract
Precisely modulating the kinetics of toehold-mediated DNA strand displacements (TMSD) is essential for its application in DNA nanotechnology. The sequence in the toehold region significantly influences the kinetics of TMSD. However, due to the large sample space resulting from various arrangements of base sequences and the resulted complex secondary structures, such a correlation is not intuitive. Herein, machine learning was employed to reveal the relationship between the kinetics of TMSD and the toehold sequence as well as the correlated secondary structure of invader strands. Key factors that influence the rate constant of TMSD were identified, such as the number of free hydrogen bonding sites in the invader, the number of free bases in the toehold, and the number of hydrogen bonds in intermediates. Moreover, a predictive model was constructed, which successfully achieved semi-quantitative prediction of rate constants of TMSD even with subtle distinctions in toehold sequence.
Collapse
Affiliation(s)
- Da Long
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Peichen Shi
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Xin Xu
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Jiayi Ren
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Yuqing Chen
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Shihui Guo
- School of Informatics, Xiamen University, Xiamen 361005, PR China
| | - Xinchang Wang
- School of Electronic Science and Engineering (National Model Microelectronics College), Xiamen University, Xiamen 361005, PR China
| | - Xiaoyu Cao
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Liulin Yang
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| | - Zhongqun Tian
- State Key Laboratory of Physical Chemistry of Solid Surface, Key Laboratory of Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials (iChEM), Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, PR China
| |
Collapse
|
50
|
Shi R, Chen KL, Fern J, Deng S, Liu Y, Scalise D, Huang Q, Cowan NJ, Gracias DH, Schulman R. Programming gel automata shapes using DNA instructions. Nat Commun 2024; 15:7773. [PMID: 39237499 PMCID: PMC11377784 DOI: 10.1038/s41467-024-51198-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/31/2024] [Indexed: 09/07/2024] Open
Abstract
The ability to transform matter between numerous physical states or shapes without wires or external devices is a major challenge for robotics and materials design. Organisms can transform their shapes using biomolecules carrying specific information and localize at sites where transitions occur. Here, we introduce gel automata, which likewise can transform between a large number of prescribed shapes in response to a combinatorial library of biomolecular instructions. Gel automata are centimeter-scale materials consisting of multiple micro-segments. A library of DNA activator sequences can each reversibly grow or shrink different micro-segments by polymerizing or depolymerizing within them. We develop DNA activator designs that maximize the extent of growth and shrinking, and a photolithography process for precisely fabricating gel automata with elaborate segmentation patterns. Guided by simulations of shape change and neural networks that evaluate gel automata designs, we create gel automata that reversibly transform between multiple, wholly distinct shapes: four different letters and every even or every odd numeral. The sequential and repeated metamorphosis of gel automata demonstrates how soft materials and robots can be digitally programmed and reprogrammed with information-bearing chemical signals.
Collapse
Affiliation(s)
- Ruohong Shi
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Kuan-Lin Chen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Joshua Fern
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Siming Deng
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Laboratory for Computational Sensing and Robotics (LCSR), Johns Hopkins University, Baltimore, MD, USA
| | - Yixin Liu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Dominic Scalise
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, USA
| | - Qi Huang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Noah J Cowan
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Laboratory for Computational Sensing and Robotics (LCSR), Johns Hopkins University, Baltimore, MD, USA
| | - David H Gracias
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Center for MicroPhysiological Systems (MPS), Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA.
| | - Rebecca Schulman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Laboratory for Computational Sensing and Robotics (LCSR), Johns Hopkins University, Baltimore, MD, USA.
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA.
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|