1
|
Zeng H, Zeng W, Liang Y. Application of multivalent aptamers in tumor diagnosis, analysis and therapy (Review). Oncol Lett 2025; 30:325. [PMID: 40370647 PMCID: PMC12076044 DOI: 10.3892/ol.2025.15071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
Cancer remains one of the leading causes of mortality worldwide, making early diagnosis and precise treatment crucial for enhancing patient survival rates. Previously, nucleic acid aptamers have emerged as promising tools in tumor diagnosis, analysis and therapy, owing to their high specificity, cost-effectiveness and ease of modification. Unlike monovalent aptamers, multivalent aptamers markedly improve target affinity and specificity through multipoint binding, demonstrating superior efficacy in the detection, capture and treatment of tumor cells. The present study reviews the construction methods of multivalent aptamers, their applications in tumor diagnosis and therapy and their challenges and prospects. Constructed through nucleic acid nanostructure self-assembly, bio-coupling, nanomaterial loading and chemical cross-linking, multivalent aptamers can effectively detect circulating tumor cells, enabling the non-invasive release of tumor cells. They serve as alternatives to bispecific antibodies in tumor immunotherapy. Despite challenges in specificity, in vivo delivery efficiency and large-scale production in the tumor microenvironment, multivalent aptamers hold promise for future research in dynamic response technology, artificial intelligence-driven development and clinical application. The present systematic review summarizes advancements in multivalent aptamers in oncology over the past decade and their potential application in precision medicine.
Collapse
Affiliation(s)
- Hongyue Zeng
- Department of Oncology, The Second Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Wenjing Zeng
- Department of Oncology, The Second Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yaoyao Liang
- Department of Oncology, The Second Affiliated Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
2
|
Zhang X, Yang Y, Tian Z, Du Z, Zhou W, Fu T, Zheng L, Luo C, Peng R, Tan W. Programmable Loading of a Multivalent LRPPRC Aptamer onto a Rectangular DNA Tile Inhibits the Proliferation of Lung Adenocarcinoma Cells. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23722-23730. [PMID: 40223205 DOI: 10.1021/acsami.5c02782] [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: 04/15/2025]
Abstract
Since cancer biomarkers for lung adenocarcinoma can lead to early intervention and treatment, they have been the focus of much research attention. DNA aptamers, which are functional oligonucleotides, exhibit high specificity and binding affinity to different types of cancer biomarkers. Through DNA aptamer screening, a leucine-rich PPR-motif-containing protein (LRPPRC) was discovered as a potential biomarker for lung adenocarcinoma therapeutics. It is an RNA-binding protein that helps in regulating post-transcriptional gene expression in mitochondria. Interestingly, the first LRPPRC-targeted small-molecule drug showed significant antitumor effects. Apart from biomarker discovery, DNA aptamers have also shown promise in cancer therapeutics, but challenges in the programmable delivery of aptamers have limited applications. Herein, we have addressed these challenges in two steps. First, after obtaining purified protein LRPPRC, we verified aptamer R14 as its high-affinity binding ligand. Second, for programmable delivery, a rectangular DNA tile (RDT) was constructed to improve cellular internalization. In particular, DNA handles on the surface of this DNA nanostructure serve as overhangs for loading multivalent R14, and both A549 and PC9 cells treated with R14-RDT targeted to LRPPRC showed significant inhibition of cancer cell proliferation. We then investigated the molecular mechanism(s) underlying the interaction between multivalent aptamer R14 loaded on an RDT and its cognate target protein such that the result is inhibition of cancer cell proliferation. Based on our findings, we hypothesized that R14-RDT-LRPPRC interaction triggers significant gene transcription and RNA processing events that result in inhibiting mitochondria-related genes and RNA transcriptional processing, while causing an immune inflammatory response that ultimately leads to the inhibition of cancer cell proliferation. Therefore, this research offers an instructive paradigm for programmable loading of a multivalent aptamer onto a two-dimensional DNA nanostructure to improve targeted cancer therapeutics through intervening with the cell's transcriptome.
Collapse
Affiliation(s)
- Xinna Zhang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, P. R. China
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Yunben Yang
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Zhan Tian
- Beijing National Research Center for Molecular Sciences, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, P. R. China
| | - Ziyan Du
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Wei Zhou
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Ting Fu
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Linfeng Zheng
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Cong Luo
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Ruizi Peng
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Weihong Tan
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, P. R. China
| |
Collapse
|
3
|
Hayat M, Bukhari SAR, Raza M, Rafia, Aslam A, Liu Z. Nanostructured aptasensors for ricin detection and tumor therapy: exploring aptamer-protein interactions and conformational stability in biological complexities. Int J Biol Macromol 2025; 310:143282. [PMID: 40254195 DOI: 10.1016/j.ijbiomac.2025.143282] [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: 03/12/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Aptamers are distinctive single-stranded oligonucleotides derived through in vitro evolution, and exhibit exceptional ability in binding to target proteins. Structural modifications of aptamers can profoundly regulate their interactions with proteins, thereby influencing associated cellular behavior. Recent research focused on modulating aptamer-protein interaction in complex biological environments to regulate various biological processes. However, in such crowded conditions, aptamer conformation and stability are susceptible to nuclease degradation, which can impair stable binding to target. Ricin is recognized as a significant biological toxin protein, distinguished by its widespread availability, remarkable dissemination, and resilience including wide pH tolerance, remarkable thermostability, and solubility. RTA is an enzymatic subunit of ricin, that can inactivate approximately 2000 ribosomes per minute, rapidly halting protein synthesis, making it a powerful candidate for tumor therapy. By leveraging the potent cytotoxicity of ricin, coupled with the targeting precision of aptamers and the versatility of nanomaterials, a powerful approach emerges for both targeted tumor therapy and highly sensitive detection of ricin. Although there have been some insightful reports on aptamers applied in ricin detection, a systematic discussion remains limited. In this context, we provide an in-depth overview of techniques used to analyze aptamer-ricin interactions and explore the potential of ricin-aptamer interactions in clinical diagnosis.
Collapse
Affiliation(s)
- Minahil Hayat
- School of Life Sciences, Shanghai University, Shanghai, China
| | | | - Mohsan Raza
- College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Rafia
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Arooj Aslam
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Zhanmin Liu
- School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
4
|
Liu X, Zhang X, Wei D, Liu Z, Yang L. Innovative bioinspired hydrogel scaffolds enabling in-situ hybrid nanoflower integration for dual-mode acetylcholinesterase inhibitor profiling. Biosens Bioelectron 2025; 271:117032. [PMID: 39665919 DOI: 10.1016/j.bios.2024.117032] [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: 10/08/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
This study introduces an innovative bioinspired hydrogel scaffold tailored to facilitate the in-situ integration of hybrid nanoflowers (HNFs) into the sensing interface, thereby establishing a versatile dual-mode platform for the sensitive profiling of acetylcholinesterase (AChE) inhibitors, a pivotal aspect in the pursuit of Alzheimer's disease therapeutics. Mimicking the tenacious anchoring of natural tree roots, our design employs magnetic bead imprinting with Strep-Tactin-coated magnetic beads (STMBs) to shape the hydrogel, which is then complemented by the integration of AChE-specific aptamers. This configuration creates a stable and biomimetic environment that nurtures HNF growth, thereby enhancing the binding integrity of HNFs with sensing interfaces. The platform's dual-mode detection capability, integrating both colorimetric and electrochemical sensing, is demonstrated through the effective evaluation of galantamine's inhibitory potency, with IC50 values that highlight its therapeutic potential. The hydrogel's exceptional reusability, maintaining over 95% of its initial activity after multiple uses, and its long-term stability, retaining 91% of its initial performance, further highlight its practicality and cost-effectiveness. In summary, this bioinspired hydrogel scaffold offers a novel, efficient, and dependable biosensing strategy for HNF-based biosensors, showing great potential for broad applications in medical diagnostics and advanced biosensing technologies.
Collapse
Affiliation(s)
- Xingyi Liu
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, PR China
| | - Xiaolong Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, PR China
| | - Dongsheng Wei
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, PR China
| | - Zhen Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, 518055, PR China.
| | - Limin Yang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, PR China.
| |
Collapse
|
5
|
Xia C, Chen X, Jiang YB, Jiang T. Self-Assembled Peptide Sheet-Mediated Multivalent Capture of Cells with Enhanced Tunability. Chembiochem 2025; 26:e202400797. [PMID: 39622775 DOI: 10.1002/cbic.202400797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/30/2024] [Indexed: 12/13/2024]
Abstract
We report the creation of multivalent ligand surfaces for cell capture by conjugation of ligand-appended 2D peptide assemblies on an antifouling glass substrate. The sheet-like structures organize ligands into non-uniform, patchy patterns, enhancing multivalent cell targeting. A 155 % increase in captured cells was achieved compared to the presentation of the ligands on surfaces lacking the peptide sheets. Being orthogonal to the commonly used dendrimer- and cyclic organic molecular-based scaffolds, this peptide assembly-based approach offers a facile method to modulate the identity, number, and spatial distribution of ligands through controlled peptide coassembly. Using this method, we constructed a surface bearing two types of ligands, which demonstrates a 128 % enhancement in targeting selectivity between two model cells compared to the mono-ligand surface. These findings illustrate that integration of peptide assemblies into ligand substrates permits robust and effective manipulation of multivalent cell targeting, advancing the development of customizable cell-binding materials.
Collapse
Affiliation(s)
- Cai Xia
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen, 361005, China
| | - Xin Chen
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen, 361005, China
- College of Materials Science and Engineering, Huaqiao University, Key Laboratory of Molecular Designing and Green Conversions (Fujian University), Xiamen, 361021, China
| | - Yun-Bao Jiang
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen, 361005, China
| | - Tao Jiang
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Xiamen, 361005, China
| |
Collapse
|
6
|
Zhou KXT, Bujold KE. The Emergence of Oligonucleotide Building Blocks in the Multispecific Proximity-Inducing Drug Toolbox of Destruction. ACS Chem Biol 2025; 20:3-18. [PMID: 39704048 DOI: 10.1021/acschembio.4c00311] [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: 12/21/2024]
Abstract
Oligonucleotides are a rapidly emerging class of therapeutics. Their most well-known examples are informational drugs that modify gene expression by binding mRNA. Despite inducing proximity between biological machinery and mRNA when applied to modulating gene expression, oligonucleotides are not typically labeled as "proximity-inducing" in literature. Yet, they have recently been explored as building blocks for multispecific proximity-inducing drugs (MPIDs). MPIDs are unique because they can direct endogenous biological machinery to destroy targeted molecules and cells, in contrast to traditional drugs that inhibit only their functions. The unique mechanism of action of MPIDs has enabled the targeting of previously "undruggable" molecular entities that cannot be effectively inhibited. However, the development of MPIDs must ensure that these molecules will selectively direct a potent, destruction-based mechanism of action toward intended targets over healthy tissues to avoid causing life-threatening toxicities. Oligonucleotides have emerged as promising building blocks for the design of MPIDs because they are sequence-controlled molecules that can be rationally designed to program multispecific binding interactions. In this Review, we examine the emergence of oligonucleotide-containing MPIDs in the proximity induction space, which has been dominated by antibody and small molecule MPID modalities. Moreover, examples of oligonucleotides developed as MPID candidates in immunotherapy and protein degradation are discussed to demonstrate the utility of oligonucleotides in expanding the scope and selectivity of the MPID toolbox. Finally, we discuss the utility of programming "AND" gates into oligonucleotide scaffolds to encode conditional responses that have the potential to be incorporated into MPIDs, which can further enhance their selectivity, thus increasing the scope of this drug category.
Collapse
Affiliation(s)
- Kevin Xiao Tong Zhou
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| | - Katherine E Bujold
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| |
Collapse
|
7
|
Luo L, Li J, Zhou Y, Xiang D, Luan Y, Wang Q, Huang J, Liu J, Yang X, Wang K. Spatially Controlled DNA Frameworks for Sensitive Detection and Specific Isolation of Tumor Cells. Angew Chem Int Ed Engl 2024; 63:e202411382. [PMID: 39405000 DOI: 10.1002/anie.202411382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/14/2024] [Indexed: 11/13/2024]
Abstract
High-affinity, specific, and sensitive probes are crucial for the specific recognition and identification of tumor cells from complex matrices. Multivalent binding is a powerful strategy, but the irrational spatial distribution of the functional moieties may reduce the probe performance. Here, we constructed a Janus DNA triangular prism nanostructure (3Zy1-JTP-3) for sensitive detection and specific isolation of tumor cells. Benefiting from spatial features of the triangular prism, the fluorescence intensity induced by 3Zy1-JTP-3 was almost 4 times that of the monovalent structure. Moreover, the DNA triangular prisms were connected to form hand-in-hand multivalent DNA triangular prism structures (Zy1-MTP), in which the fluorescence intensity and affinity were increased to 9-fold and 10-fold of 3Zy1-JTP-3, respectively. Furthermore, 3Zy1-JTP-3 and Zy1-MTP were combined with magnetic beads, and the latter showed higher capture efficiency (>90 %) in whole blood. This work provides a new strategy for the efficient capture of rare cells in complex biological samples.
Collapse
Affiliation(s)
- Lei Luo
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Jiaojiao Li
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yuan Zhou
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Dongliu Xiang
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Yanan Luan
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Qing Wang
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - 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, China
| | - Jianbo Liu
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Xiaohai Yang
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| | - Kemin Wang
- State key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, China
| |
Collapse
|
8
|
Leng G, Duan B, Liu J, Li S, Zhao W, Wang S, Hou G, Qu J. The advancements and prospective developments in anti-tumor targeted therapy. Neoplasia 2024; 56:101024. [PMID: 39047659 PMCID: PMC11318541 DOI: 10.1016/j.neo.2024.101024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Cancer poses a major threat to human health worldwide. The development of anti-tumor materials provides new modalities for cancer diagnosis and treatment. In this review, we comprehensively summarize the research progress and clinical applications of anti-tumor materials. First, we introduce the etiology and pathogenesis of cancer, and the significance and challenges of anti-tumor materials research. Then, we classify anti-tumor materials and discuss their mechanisms of action. After that, we elaborate the research advances and clinical applications of anti-tumor materials, including those targeting tumor cells and therapeutic instruments. Finally, we discuss the future perspectives and challenges in the field of anti-tumor materials. This review aims to provide an overview of the current status of anti-tumor materials research and application, and to offer insights into future directions in this rapidly evolving field, which holds promise for more precise, efficient and customized treatment of cancer.
Collapse
Affiliation(s)
- Guorui Leng
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Baorong Duan
- Research Center for Leather and Protein of College of Chemistry & Chemical Engineering, Yantai University, Yantai 264005, China
| | - Junjie Liu
- Department of Physics, Binzhou Medical University, Yantai 264003, China
| | - Song Li
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Wenwen Zhao
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Shanshan Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Guige Hou
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| | - Jiale Qu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China; Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS); Shandong Provincial Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, China.
| |
Collapse
|
9
|
Hamidi SV, Jahromi AK, Hosseini II, Moakhar RS, Collazos C, Pan Q, Liang C, Mahshid S. Surface-Based Multimeric Aptamer Generation and Bio-Functionalization for Electrochemical Biosensing Applications. Angew Chem Int Ed Engl 2024; 63:e202402808. [PMID: 38764376 DOI: 10.1002/anie.202402808] [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: 02/07/2024] [Revised: 05/05/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
Multimeric aptamers have gained more attention than their monomeric counterparts due to providing more binding sites for target analytes, leading to increased affinity. This work attempted to engineer the surface-based generation of multimeric aptamers by employing the room temperature rolling circle amplification (RCA) technique and chemically modified primers for developing a highly sensitive and selective electrochemical aptasensor. The multimeric aptamers, generated through surface RCA, are hybridized to modified spacer primers, facilitating the positioning of the aptamers in the proximity of sensing surfaces. These multimeric aptamers can be used as bio-receptors for capturing specific targets. The surface amplification process was fully characterized, and the optimal amplification time for biosensing purposes was determined, using SARS-CoV-2 spike protein (SP). Interestingly, multimeric aptasensors produced considerably higher response signals and affinity (more than 10-fold), as well as higher sensitivity (almost 4-fold) compared to monomeric aptasensors. Furthermore, the impact of surface structures on the response signals was studied by utilizing both flat working electrodes (WEs) and nano-/microislands (NMIs) WEs. The NMIs multimeric aptasensors showed significantly higher sensitivity in buffer and saliva media with the limit of detection less than 2 fg/ml. Finally, the developed NMIs multimeric aptasensors were clinically challenged with several saliva patient samples.
Collapse
Affiliation(s)
- Seyed Vahid Hamidi
- Department of Bioengineering, McGill University, Montreal, Quebec, H3A 0E9, Canada
| | | | - Imman I Hosseini
- Department of Bioengineering, McGill University, Montreal, Quebec, H3A 0E9, Canada
| | | | - Cesar Collazos
- Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
- Lady Davis Institute for Medical Research and McGill Centre for Viral Diseases, Jewish General Hospital, Montreal, Quebec, 3T 1E2, Canada
| | - Qinghua Pan
- Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
- Lady Davis Institute for Medical Research and McGill Centre for Viral Diseases, Jewish General Hospital, Montreal, Quebec, 3T 1E2, Canada
| | - Chen Liang
- Department of Medicine, McGill University, Montreal, Quebec, H4A 3J1, Canada
- Lady Davis Institute for Medical Research and McGill Centre for Viral Diseases, Jewish General Hospital, Montreal, Quebec, 3T 1E2, Canada
| | - Sara Mahshid
- Department of Bioengineering, McGill University, Montreal, Quebec, H3A 0E9, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, H3A 0G4, Canada
| |
Collapse
|
10
|
Kawamoto Y, Wu Y, Park S, Hidaka K, Sugiyama H, Takahashi Y, Takakura Y. Multivalent dendritic DNA aptamer molecules for the enhancement of therapeutic effects. Chem Commun (Camb) 2024; 60:6256-6259. [PMID: 38768325 DOI: 10.1039/d4cc00578c] [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: 05/22/2024]
Abstract
Dendritic DNA molecules, referred to as DNA dendrons, consist of multiple covalently linked strands and are expected to improve the cellular uptake and potency of therapeutic oligonucleotides because of their multivalency. In this study, we developed an efficient synthetic method for producing DNA dendrons using strain-promoted azide-alkyne cycloaddition. Integration of the antitumor aptamer AS1411 into DNA dendrons enhanced cellular uptake and antiproliferative activity in cancer cells. These findings demonstrate that the incorporation of multivalent aptamers into DNA dendrons can effectively boost their therapeutic effects.
Collapse
Affiliation(s)
- Yusuke Kawamoto
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - You Wu
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Soyoung Park
- Immunology Frontier Research Center, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kumi Hidaka
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hiroshi Sugiyama
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Yoshida-ushinomiyacho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
11
|
Ye T, Xu Y, Chen H, Yuan M, Cao H, Hao L, Wu X, Yin F, Xu F. A trivalent aptasensor by using DNA tetrahedron as scaffold for label-free determination of antibiotics. Biosens Bioelectron 2024; 251:116127. [PMID: 38382272 DOI: 10.1016/j.bios.2024.116127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/09/2024] [Accepted: 02/11/2024] [Indexed: 02/23/2024]
Abstract
Owing to advantage in high sensitivity and fast response, aptamer based electrochemical biosensors have attracted much more attention. However, inappropriate interfacial engineering strategy leads to poor recognition performance, which ascribe to the following factors of immobilized oligonucleotide strand including steric hindrance, interchain entanglement, and unfavorable conformation. In this work, we proposed a DNA tetrahedron based diblock aptamer immobilized strategy for the construction of label-free electrochemical biosensor. The diblock aptamer sequence is composite of T-rich anchor domain and recognition domain, where T-rich domain enabling anchored on the edge of DNA tetrahedron via Hoogsteen hydrogen bond at neutral condition. The DNA tetrahedron scaffold offers an appropriate lateral space for target recognition of diblock aptamer. More importantly, this trivalent aptamer recognition interface can be regenerated by simply adjusting the pH environment to alkaline, resulting in the dissociation of diblock aptamer. Under the optimum condition, proposed electrochemical aptasensor manifested a satisfied sensitivity for aminoglycosides antibiotic, kanamycin with a limit of detection of 0.69 nM, which is 45-fold lower than traditional Au-S immobilization strategy. Moreover, the proposed aptasensor had also successfully been extended to ampicillin detection by changing the sequence of recognition domain in diblock aptamer. This work paves a new way for the rational design of aptamer-based electrochemical sensor.
Collapse
Affiliation(s)
- Tai Ye
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yimin Xu
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Haohao Chen
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Min Yuan
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Hui Cao
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Liling Hao
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Xiuxiu Wu
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Fengqin Yin
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Fei Xu
- Shanghai Engineering Research Center of Food Rapid Detection, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| |
Collapse
|
12
|
Li X, Li G, Pan Q, Xue F, Wang Z, Peng C. Rapid and ultra-sensitive lateral flow assay for pathogens based on multivalent aptamer and magnetic nanozyme. Biosens Bioelectron 2024; 250:116044. [PMID: 38271888 DOI: 10.1016/j.bios.2024.116044] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/03/2024] [Accepted: 01/13/2024] [Indexed: 01/27/2024]
Abstract
Ultra-sensitive LFA methods for pathogen detection commonly depended on tedious and time-consuming nucleic acid amplification. Here, a high affinity multivalent aptamer (multi-Apt) for S. aureus was obtained through exquisite engineering design. The scaffold and conformation of the multi-Apt were found to be key factors in the detection signal of aptsensors. After optimization, the binding affinity of the multi-Apt to S. aureus was improved by more than 8-fold from 135.9 nM to 16.77 nM. By the joint use of the multi-Apt and a multifunctional nanozyme Fe3O4@MOF@PtPd, a fast and ultra-sensitive LFA for S. aureus was developed (termed MA-MN LFA). In this method, a Fe3O4@MOF@PtPd nanozyme was modified with vancomycin and could efficiently capture and separate S. aureus. Moreover, the multi-Apt worked together with the nanozyme to bind with S. aureus to form a ternary complex at the same time, which simply the fabrication of LFA strip. The developed MA-MN LFA could detect S. aureus as low as 2 CFU/mL within 30 min and a wide linear range of 10-1 × 108 CFU/mL was obtained. The detection is easily operated, fast (can be completed within 30 min) and versatile for Gram-positive pathogens, thus has great potential as a powerful tool in pathogen detection.
Collapse
Affiliation(s)
- Xiuping Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China
| | - Guowen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China
| | - Qiuli Pan
- Shandong Institute for Food and Drug Control, Jinan 250101, PR China
| | - Feng Xue
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhouping Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China; International Joint Laboratory on Food Safety, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China
| | - Chifang Peng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China; International Joint Laboratory on Food Safety, Jiangnan University, Lihu Road 1800, Wuxi 214122, PR China.
| |
Collapse
|
13
|
Wang GY, Yan DX, Rong RX, Shi BY, Lin GJ, Yin F, Wei WT, Li XL, Wang KR. Amphiphilic α-Peptoid-deoxynojirimycin Conjugate-based Multivalent Glycosidase Inhibitor for Hypoglycemic Effect and Fluorescence Imaging. J Med Chem 2024; 67:5945-5956. [PMID: 38504504 DOI: 10.1021/acs.jmedchem.4c00304] [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: 03/21/2024]
Abstract
Multivalent glycosidase inhibitors based on 1-deoxynojirimycin derivatives against α-glucosidases have been rapidly developed. Nonetheless, the mechanism based on self-assembled multivalent glucosidase inhibitors in living systems needs to be further studied. It remains to be determined whether the self-assembly possesses sufficient stability to endure transit through the small intestine and subsequently bind to the glycosidases located therein. In this paper, two amphiphilic compounds, 1-deoxynojirimycin and α-peptoid conjugates (LP-4DNJ-3C and LP-4DNJ-6C), were designed. Their self-assembling behaviors, multivalent α-glucosidase inhibition effect, and fluorescence imaging on living organs were studied. LP-4DNJ-6C exhibited better multivalent α-glucosidase inhibition activities in vitro. Moreover, the self-assembly of LP-4DNJ-6C could effectively form a complex with Nile red. The complex showed fluorescence quenching effect upon binding with α-glucosidases and exhibited potent fluorescence imaging in the small intestine. This result suggests that a multivalent hypoglycemic effect achieved through self-assembly in the intestine is a viable approach, enabling the rational design of multivalent hypoglycemic drugs.
Collapse
Affiliation(s)
- Guang-Yuan Wang
- College of Chemistry and Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding 071002, P. R. China
- College of Chemical Engineering & Material, Hebei Key Laboratory of Heterocyclic Compounds, Handan Key Laboratory of Organic Small Molecule Materials, Handan University, Handan 056005, P. R. China
| | - Dong-Xiao Yan
- Department of Immunology, School of Basic Medical Science, Hebei University, Baoding 071002, P. R. China
| | - Rui-Xue Rong
- Department of Immunology, School of Basic Medical Science, Hebei University, Baoding 071002, P. R. China
| | - Bing-Ye Shi
- Affiliated Hospital of Hebei University, Hebei University, Baoding 071002, P. R. China
| | - Gao-Juan Lin
- College of Chemistry and Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding 071002, P. R. China
| | - Fangqian Yin
- College of Chemistry and Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding 071002, P. R. China
- College of Chemical Engineering & Material, Hebei Key Laboratory of Heterocyclic Compounds, Handan Key Laboratory of Organic Small Molecule Materials, Handan University, Handan 056005, P. R. China
| | - Wen-Tong Wei
- College of Chemistry and Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding 071002, P. R. China
| | - Xiao-Liu Li
- College of Chemistry and Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding 071002, P. R. China
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Key Laboratory of Chemical Biology of Hebei Province, Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding 071002, P. R. China
| |
Collapse
|
14
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
15
|
Ghosh S, Ghosh S, Sharma H, Bhaskar R, Han SS, Sinha JK. Harnessing the power of biological macromolecules in hydrogels for controlled drug release in the central nervous system: A review. Int J Biol Macromol 2024; 254:127708. [PMID: 37923043 DOI: 10.1016/j.ijbiomac.2023.127708] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
Hydrogels have immense potential in revolutionizing central nervous system (CNS) drug delivery, improving outcomes for neurological disorders. They serve as promising tools for controlled drug delivery to the CNS. Available hydrogel types include natural macromolecules (e.g., chitosan, hyaluronic acid, alginate), as well as hybrid hydrogels combining natural and synthetic polymers. Each type offers distinct advantages in terms of biocompatibility, mechanical properties, and drug release kinetics. Design and engineering considerations encompass hydrogel composition, crosslinking density, porosity, and strategies for targeted drug delivery. The review emphasizes factors affecting drug release profiles, such as hydrogel properties and formulation parameters. CNS drug delivery applications of hydrogels span a wide range of therapeutics, including small molecules, proteins and peptides, and nucleic acids. However, challenges like limited biodegradability, clearance, and effective CNS delivery persist. Incorporating 3D bioprinting technology with hydrogel-based CNS drug delivery holds the promise of highly personalized and precisely controlled therapeutic interventions for neurological disorders. The review explores emerging technologies like 3D bioprinting and nanotechnology as opportunities for enhanced precision and effectiveness in hydrogel-based CNS drug delivery. Continued research, collaboration, and technological advancements are vital for translating hydrogel-based therapies into clinical practice, benefiting patients with CNS disorders. This comprehensive review article delves into hydrogels for CNS drug delivery, addressing their types, design principles, applications, challenges, and opportunities for clinical translation.
Collapse
Affiliation(s)
- Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India; ICMR - National Institute of Nutrition, Tarnaka, Hyderabad, Telangana 500007, India
| | - Soumya Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Hitaishi Sharma
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea.
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea.
| | | |
Collapse
|
16
|
Pan W, Tao T, Qiu Y, Zhu X, Zhou X. Natural killer cells at the forefront of cancer immunotherapy with immune potency, genetic engineering, and nanotechnology. Crit Rev Oncol Hematol 2024; 193:104231. [PMID: 38070841 DOI: 10.1016/j.critrevonc.2023.104231] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Natural killer (NK) cells are vital components of the human immune system, acting as innate lymphocytes and playing a crucial role in immune surveillance. Their unique ability to independently eliminate target cells without antigen contact or antibodies has sparked interest in immunological research. This review examines recent NK cell developments and applications, encompassing immune functions, interactions with target cells, genetic engineering techniques, pharmaceutical interventions, and implications in cancers. Insights into NK cell regulation emerge, with a focus on promising genetic engineering like CAR-engineered NK cells, enhancing specificity against tumors. Immune checkpoint inhibitors also enhance NK cells' potential in cancer therapy. Nanotechnology's emergence as a tool for targeted drug delivery to improve NK cell therapies is explored. In conclusion, NK cells are pivotal in immunity, holding exciting potential in cancer immunotherapy. Ongoing research promises novel therapeutic strategies, advancing immunotherapy and medical interventions.
Collapse
Affiliation(s)
- Weiyi Pan
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; School of Public Health, Southern Medical University, Guangzhou, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Yishu Qiu
- Department of Biology, College of Arts and Science, New York University, New York, USA
| | - Xiao Zhu
- Computational Systems Biology Lab (CSBL), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
17
|
Almeida‐Pinto J, Lagarto MR, Lavrador P, Mano JF, Gaspar VM. Cell Surface Engineering Tools for Programming Living Assemblies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304040. [PMID: 37823678 PMCID: PMC10700290 DOI: 10.1002/advs.202304040] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/10/2023] [Indexed: 10/13/2023]
Abstract
Breakthroughs in precision cell surface engineering tools are supporting the rapid development of programmable living assemblies with valuable features for tackling complex biological problems. Herein, the authors overview the most recent technological advances in chemically- and biologically-driven toolboxes for engineering mammalian cell surfaces and triggering their assembly into living architectures. A particular focus is given to surface engineering technologies for enabling biomimetic cell-cell social interactions and multicellular cell-sorting events. Further advancements in cell surface modification technologies may expand the currently available bioengineering toolset and unlock a new generation of personalized cell therapeutics with clinically relevant biofunctionalities. The combination of state-of-the-art cell surface modifications with advanced biofabrication technologies is envisioned to contribute toward generating living materials with increasing tissue/organ-mimetic bioactivities and therapeutic potential.
Collapse
Affiliation(s)
- José Almeida‐Pinto
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Matilde R. Lagarto
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Pedro Lavrador
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - João F. Mano
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Vítor M. Gaspar
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| |
Collapse
|
18
|
Liu C, Hu L, Dong G, Zhang Y, Ferreira da Silva-Júnior E, Liu X, Menéndez-Arias L, Zhan P. Emerging drug design strategies in anti-influenza drug discovery. Acta Pharm Sin B 2023; 13:4715-4732. [PMID: 38045039 PMCID: PMC10692392 DOI: 10.1016/j.apsb.2023.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/12/2023] [Accepted: 08/03/2023] [Indexed: 12/05/2023] Open
Abstract
Influenza is an acute respiratory infection caused by influenza viruses (IFV), According to the World Health Organization (WHO), seasonal IFV epidemics result in approximately 3-5 million cases of severe illness, leading to about half a million deaths worldwide, along with severe economic losses and social burdens. Unfortunately, frequent mutations in IFV lead to a certain lag in vaccine development as well as resistance to existing antiviral drugs. Therefore, it is of great importance to develop anti-IFV drugs with high efficiency against wild-type and resistant strains, needed in the fight against current and future outbreaks caused by different IFV strains. In this review, we summarize general strategies used for the discovery and development of antiviral agents targeting multiple IFV strains (including those resistant to available drugs). Structure-based drug design, mechanism-based drug design, multivalent interaction-based drug design and drug repurposing are amongst the most relevant strategies that provide a framework for the development of antiviral drugs targeting IFV.
Collapse
Affiliation(s)
- Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Lide Hu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Guanyu Dong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Ying Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Edeildo Ferreira da Silva-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió 57072-970, Alagoas, Brazil
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Luis Menéndez-Arias
- Centro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid), Madrid 28049, Spain
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
19
|
Ji C, Wei J, Zhang L, Hou X, Tan J, Yuan Q, Tan W. Aptamer-Protein Interactions: From Regulation to Biomolecular Detection. Chem Rev 2023; 123:12471-12506. [PMID: 37931070 DOI: 10.1021/acs.chemrev.3c00377] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Serving as the basis of cell life, interactions between nucleic acids and proteins play essential roles in fundamental cellular processes. Aptamers are unique single-stranded oligonucleotides generated by in vitro evolution methods, possessing the ability to interact with proteins specifically. Altering the structure of aptamers will largely modulate their interactions with proteins and further affect related cellular behaviors. Recently, with the in-depth research of aptamer-protein interactions, the analytical assays based on their interactions have been widely developed and become a powerful tool for biomolecular detection. There are some insightful reviews on aptamers applied in protein detection, while few systematic discussions are from the perspective of regulating aptamer-protein interactions. Herein, we comprehensively introduce the methods for regulating aptamer-protein interactions and elaborate on the detection techniques for analyzing aptamer-protein interactions. Additionally, this review provides a broad summary of analytical assays based on the regulation of aptamer-protein interactions for detecting biomolecules. Finally, we present our perspectives regarding the opportunities and challenges of analytical assays for biological analysis, aiming to provide guidance for disease mechanism research and drug discovery.
Collapse
Affiliation(s)
- 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
| | - Junyuan Wei
- 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
| | - Lei 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
| | - Xinru Hou
- 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
| | - 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
| | - 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
| | - Weihong 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
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
20
|
Szymanowski W, Szymanowska A, Bielawska A, Lopez-Berestein G, Rodriguez-Aguayo C, Amero P. Aptamers as Potential Therapeutic Tools for Ovarian Cancer: Advancements and Challenges. Cancers (Basel) 2023; 15:5300. [PMID: 37958473 PMCID: PMC10647731 DOI: 10.3390/cancers15215300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Ovarian cancer (OC) is the most common lethal gynecologic cause of death in women worldwide, with a high mortality rate and increasing incidence. Despite advancements in the treatment, most OC patients still die from their disease due to late-stage diagnosis, the lack of effective diagnostic methods, and relapses. Aptamers, synthetic, short single-stranded oligonucleotides, have emerged as promising anticancer therapeutics. Their ability to selectively bind to target molecules, including cancer-related proteins and receptors, has revolutionized drug discovery and biomarker identification. Aptamers offer unique insights into the molecular pathways involved in cancer development and progression. Moreover, they show immense potential as drug delivery systems, enabling targeted delivery of therapeutic agents to cancer cells while minimizing off-target effects and reducing systemic toxicity. In the context of OC, the integration of aptamers with non-coding RNAs (ncRNAs) presents an opportunity for precise and efficient gene targeting. Additionally, the conjugation of aptamers with nanoparticles allows for accurate and targeted delivery of ncRNAs to specific cells, tissues, or organs. In this review, we will summarize the potential use and challenges associated with the use of aptamers alone or aptamer-ncRNA conjugates, nanoparticles, and multivalent aptamer-based therapeutics for the treatment of OC.
Collapse
Affiliation(s)
- Wojciech Szymanowski
- Department of Biotechnology, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.S.); (A.B.)
| | - Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.S.); (A.B.)
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
| |
Collapse
|
21
|
Shang Z, Deng Z, Yi X, Yang M, Nong X, Lin M, Xia F. Construction and bioanalytical applications of poly-adenine-mediated gold nanoparticle-based spherical nucleic acids. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:5564-5576. [PMID: 37861233 DOI: 10.1039/d3ay01618h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Owing to the versatile photophysical and chemical properties, spherical nucleic acids (SNAs) have been widely used in biosensing. However, traditional SNAs are formed by self-assembly of thiolated DNA on the surface of a gold nanoparticle (AuNP), where it is challenging to precisely control the orientation and surface density of DNA. As a new SNA, a polyadenine (polyA)-mediated SNA using the high binding affinity of consecutive adenines to AuNPs shows controllable surface density and configuration of DNA, which can be used to improve the performance of a biosensor. Herein, we first introduce the properties of polyA-mediated SNAs and fundamental principles regarding the polyA-AuNP interaction. Then, we provide an overview of current representative synthesis methods of polyA-mediated SNAs and their advantages and disadvantages. After that, we summarize the application of polyA-mediated SNAs in biosensing based on fluorescence and colorimetric methods, followed by discussion and an outlook of future challenges in this field.
Collapse
Affiliation(s)
- Zhiwei Shang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China.
| | - Zixuan Deng
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China.
| | - Xiaoqing Yi
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Mengyu Yang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China.
| | - Xianliang Nong
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China.
| | - Meihua Lin
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China.
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China.
| |
Collapse
|
22
|
Ma N, Sun M, Shi H, Xue L, Zhang M, Yang W, Dang Y, Qiao Z. A Colorimetric/Fluorescent Dual-Mode Aptasensor for Salmonella Based on the Magnetic Separation of Aptamers and a DNA-Nanotriangle Programmed Multivalent Aptamer. Foods 2023; 12:3853. [PMID: 37893744 PMCID: PMC10606715 DOI: 10.3390/foods12203853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Salmonella infection has emerged as a global health threat, causing death, disability, and socioeconomic disruption worldwide. The rapid and sensitive detection of Salmonella is of great significance in guaranteeing food safety. Herein, we developed a colorimetric/fluorescent dual-mode method based on a DNA-nanotriangle programmed multivalent aptamer for the sensitive detection of Salmonella. In this system, aptamers are precisely controlled and assembled on a DNA nanotriangle structure to fabricate a multivalent aptamer (NTri-Multi-Apt) with enhanced binding affinity and specificity toward Salmonella. The NTri-Multi-Apt was designed to carry many streptavidin-HRPs for colorimetric read-outs and a large load of Sybr green I in the dsDNA scaffold for the output of a fluorescent signal. Therefore, combined with the magnetic separation of aptamers and the prefabricated NTri-Multi-Apt, the dual-mode approach achieved simple and sensitive detection, with LODs of 316 and 60 CFU/mL for colorimetric and fluorescent detection, respectively. Notably, the fluorescent mode provided a self-calibrated and fivefold-improved sensitivity over colorimetric detection. Systematic results also revealed that the proposed dual-mode method exhibited high specificity and applicability for milk, egg white, and chicken meat samples, serving as a promising tool for real bacterial sample testing. As a result, the innovative dual-mode detection method showed new insights for the detection of other pathogens.
Collapse
Affiliation(s)
- Na Ma
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| | - Mengni Sun
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| | - Hanxing Shi
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| | - Liangliang Xue
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| | - Min Zhang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| | - Wenge Yang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| | - Yali Dang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| | - Zhaohui Qiao
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315800, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo 315800, China
| |
Collapse
|
23
|
Cheng Y, Qu Z, Jiang Q, Xu T, Zheng H, Ye P, He M, Tong Y, Ma Y, Bao A. Functional Materials for Subcellular Targeting Strategies in Cancer Therapy: Progress and Prospects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2305095. [PMID: 37665594 DOI: 10.1002/adma.202305095] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/26/2023] [Indexed: 09/05/2023]
Abstract
Neoadjuvant and adjuvant therapies have made significant progress in cancer treatment. However, tumor adjuvant therapy still faces challenges due to the intrinsic heterogeneity of cancer, genomic instability, and the formation of an immunosuppressive tumor microenvironment. Functional materials possess unique biological properties such as long circulation times, tumor-specific targeting, and immunomodulation. The combination of functional materials with natural substances and nanotechnology has led to the development of smart biomaterials with multiple functions, high biocompatibilities, and negligible immunogenicities, which can be used for precise cancer treatment. Recently, subcellular structure-targeting functional materials have received particular attention in various biomedical applications including the diagnosis, sensing, and imaging of tumors and drug delivery. Subcellular organelle-targeting materials can precisely accumulate therapeutic agents in organelles, considerably reduce the threshold dosages of therapeutic agents, and minimize drug-related side effects. This review provides a systematic and comprehensive overview of the research progress in subcellular organelle-targeted cancer therapy based on functional nanomaterials. Moreover, it explains the challenges and prospects of subcellular organelle-targeting functional materials in precision oncology. The review will serve as an excellent cutting-edge guide for researchers in the field of subcellular organelle-targeted cancer therapy.
Collapse
Affiliation(s)
- Yanxiang Cheng
- Department of Gynecology, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Zhen Qu
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Qian Jiang
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Tingting Xu
- Department of Clinical Laboratory, Wuhan Blood Center (WHBC), No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Hongyun Zheng
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Mingdi He
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Yongqing Tong
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| | - Yan Ma
- Department of Blood Transfusion Research, Wuhan Blood Center (WHBC), HUST-WHBC United Hematology Optical Imaging Center, No.8 Baofeng 1st Road, Wuhan, Hubei, 430030, P. R. China
| | - Anyu Bao
- Department of Clinical Laboratory, Renmin Hospital, Wuhan University, No.238 Jiefang Road, Wuchang, Wuhan, 430060, P. R. China
| |
Collapse
|
24
|
Tan KF, In LLA, Vijayaraj Kumar P. Surface Functionalization of Gold Nanoparticles for Targeting the Tumor Microenvironment to Improve Antitumor Efficiency. ACS APPLIED BIO MATERIALS 2023; 6:2944-2981. [PMID: 37435615 DOI: 10.1021/acsabm.3c00202] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Gold nanoparticles (AuNPs) have undergone significant research for their use in the treatment of cancer. Numerous researchers have established their potent antitumor properties, which have greatly impacted the treatment of cancer. AuNPs have been used in four primary anticancer treatment modalities, namely radiation, photothermal therapy, photodynamic therapy, and chemotherapy. However, the ability of AuNPs to destroy cancer is lacking and can even harm healthy cells without the right direction to transport them to the tumor microenvironment. Consequently, a suitable targeting technique is needed. Based on the distinct features of the human tumor microenvironment, this review discusses four different targeting strategies that target the four key features of the tumor microenvironment, including abnormal vasculature, overexpression of specific receptors, an acidic microenvironment, and a hypoxic microenvironment, to direct surface-functionalized AuNPs to the tumor microenvironment and increase antitumor efficacies. In addition, some current completed or ongoing clinical trials of AuNPs will also be discussed below to further reinforce the concept of using AuNPs in anticancer therapy.
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
25
|
Ai L, Jiang X, Zhang K, Cui C, Liu B, Tan W. Tools and techniques for the discovery of therapeutic aptamers: recent advances. Expert Opin Drug Discov 2023; 18:1393-1411. [PMID: 37840268 DOI: 10.1080/17460441.2023.2264187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/25/2023] [Indexed: 10/17/2023]
Abstract
INTRODUCTION The pursuit of novel therapeutic agents for serious diseases such as cancer has been a global endeavor. Aptamers characteristic of high affinity, programmability, low immunogenicity, and rapid permeability hold great promise for the treatment of diseases. Yet obtaining the approval for therapeutic aptamers remains challenging. Consequently, researchers are increasingly devoted to exploring innovative strategies and technologies to advance the development of these therapeutic aptamers. AREAS COVERED The authors provide a comprehensive summary of the recent progress of the SELEX (Systematic Evolution of Ligands by EXponential enrichment) technique, and how the integration of modern tools has facilitated the identification of therapeutic aptamers. Additionally, the engineering of aptamers to enhance their functional attributes, such as inhibiting and targeting, is discussed, demonstrating the potential to broaden their scope of utility. EXPERT OPINION The grand potential of aptamers and the insufficient development of relevant drugs have spurred countless efforts for stimulating their discovery and application in the therapeutic field. While SELEX techniques have undergone significant developments with the aid of advanced analysis instruments and ingeniously updated aptameric engineering strategies, several challenges still impede their clinical translation. A key challenge lies in the insufficient understanding of binding conformation and susceptibility to degradation under physiological conditions. Despite the hurdles, our opinion is optimistic. With continued progress in overcoming these obstacles, the widespread utilization of aptamers for clinical therapy is envisioned to become a reality soon.
Collapse
Affiliation(s)
- Lili Ai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
| | - Xinyi Jiang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
| | - Kejing Zhang
- Department of Geriatrics and Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, The People's Republic of China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, The People's Republic of China
| | - Cheng Cui
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
| | - Bo Liu
- Department of Geriatrics and Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, The People's Republic of China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, The People's Republic of China
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, The People's Republic of China
| |
Collapse
|
26
|
Farahbakhsh Z, Zamani M, Nasirian V, Shariati L, Kermani S, Karizmeh MS, Rafienia M. An insight into fluorescence and magnetic resonance bioimaging using a multifunctional polyethyleneimine-passivated gadocarbon dots nanoconstruct assembled with AS1411. Mikrochim Acta 2023; 190:275. [PMID: 37358641 DOI: 10.1007/s00604-023-05853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/29/2023] [Indexed: 06/27/2023]
Abstract
A nanoassembly of PEI-passivated Gd@CDs, a type of aptamer, is presented which was designed and characterized in order to target specific cancer cells based on their recognition of the receptor nucleolin (NCL), which is overexpressed on the cell membrane of breast cancer cells for fluorescence and magnetic resonance imaging and treatment. Using hydrothermal methods, Gd-doped nanostructures were synthesized, then modified by a two-step chemical procedure for subsequent applications: the passivating of Gd@CDs with branched polyethyleneimine (PEI) (to form Gd@CDs-PEI1 and Gd@CDs-PEI2), and using AS1411 aptamer (AS) as a DNA-targeted molecule (to generate AS/Gd@CDs-PEI1 and AS/Gd@CDs-PEI2). Consequently, these nanoassemblies were constructed as a result of electrostatic interactions between cationic Gd@CDs-passivated PEI and AS aptamers, offering efficient multimodal targeting nanoassemblies for cancer cell detection. It has been demonstrated through in vitro studies that both types of AS-conjugated nanoassemblies are highly biocompatible, have high cellular uptake efficiency (equivalent concentration of AS: 0.25 μΜ), and enable targeted fluorescence imaging in nucleolin-positive MCF7 and MDA-MB-231 cancer cells compared to MCF10-A normal cells. Importantly, the as-prepared Gd@CDs, Gd@CDs-PEI1, and Gd@CDs-PEI2 exhibit higher longitudinal relaxivity values (r1) compared with the commercial Gd-DTPA, equal to 5.212, 7.488, and 5.667 mM-1s-1, respectively. Accordingly, it is concluded that the prepared nanoassemblies have the potential to become excellent candidates for cancer targeting and fluorescence/MR imaging agents, which can be applied to cancer imaging and personalized nanomedicine.
Collapse
Affiliation(s)
- Zohreh Farahbakhsh
- Department of Biology, Nourdanesh Institute of Higher Education, Meymeh, Isfahan, Iran
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Biosensor Research Center (BRC), Isfahan University of Medical Sciences (IUMS), Isfahan, Iran
| | - Mohammadreza Zamani
- Department of Biology, Nourdanesh Institute of Higher Education, Meymeh, Isfahan, Iran
| | - Vahid Nasirian
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Laleh Shariati
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, IR, Iran
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Kermani
- Department of Bioelectric and Biomedical Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Shie Karizmeh
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Rafienia
- Biosensor Research Center (BRC), Isfahan University of Medical Sciences (IUMS), Isfahan, Iran.
| |
Collapse
|
27
|
Liu Y, Qian X, Ran C, Li L, Fu T, Su D, Xie S, Tan W. Aptamer-Based Targeted Protein Degradation. ACS NANO 2023; 17:6150-6164. [PMID: 36942868 DOI: 10.1021/acsnano.2c10379] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The selective removal of misfolded, aggregated, or aberrantly overexpressed protein plays an essential role in maintaining protein-dominated biological processes. In parallel, the precise knockout of abnormal proteins is inseparable from the accurate identification of proteins within complex environments. Guided by these precepts, small molecules, or antibodies, are commonly used as protein recognition tools for developing targeted protein degradation (TPD) technology. Indeed, TPD has shown tremendous prospects in chronic diseases, rare diseases, cancer research, and other fields. Meanwhile, aptamers are short RNA or DNA oligonucleotides that can bind to target proteins with high specificity and strong affinity. Accordingly, aptamers are actively used in designing and constructing TPD technology. In this perspective, we provide a brief introduction to TPD technology in its current progress, and we summarize its application challenges. Recent advances in aptamer-based TPD technology are reviewed, together with corresponding challenges and outlooks.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Pathology, 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 310024, China
| | - Xu Qian
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chunyan Ran
- Department of Pathology, 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 310024, China
| | - Longjie Li
- Department of Pathology, 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 310024, China
| | - Ting Fu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Dan Su
- Department of Pathology, 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 310024, China
| | - Sitao Xie
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Department of Pathology, 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 310024, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
28
|
Han C, Xing W, Li W, Fang X, Zhao J, Ge F, Ding W, Qu P, Luo Z, Zhang L. Aptamers dimerization inspired biomimetic clamp assay towards impedimetric SARS-CoV-2 antigen detection. SENSORS AND ACTUATORS. B, CHEMICAL 2023; 380:133387. [PMID: 36694572 PMCID: PMC9851723 DOI: 10.1016/j.snb.2023.133387] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 06/17/2023]
Abstract
Antigen-detecting rapid diagnostic testing (Ag-RDT) has contributed to containing the spread of SARS-CoV-2 variants of concern (VOCs). In this study, we proposed a biomimetic clamp assay for impedimetric SARS-CoV-2 nucleocapsid protein (Np) detection. The DNA biomimetic clamp (DNA-BC) is formed by a pair of Np aptamers connected via a T20 spacer. The 5'- terminal of the DNA-BC is phosphate-modified and then anchored on the surface of the screen-printed gold electrode, which has been pre-coated with Au@UiO-66-NH2. The integrated DNA-material sensing biochip is fabricated through the strong Zr-O-P bonds to form a clamp-type impedimetric aptasensor. It is demonstrated that the aptasensor could achieve Np detection in one step within 11 min and shows pronounced sensitivity with a detection limit of 0.31 pg mL-1. Above all, the aptasensor displays great specificity and stability under physiological conditions as well as various water environments. It is a potentially promising strategy to exploit reliable Ag-RDT products to confront the ongoing epidemic.
Collapse
Affiliation(s)
- Cong Han
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Wenping Xing
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Wenjin Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Xiaona Fang
- The Cancer Hospital of the University of Chinese Academy of Sciences, Aptamer Selection Center, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jian Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Feng Ge
- Department of Gynecological Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin 300071, China
| | - Wei Ding
- Department of Gynecological Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin 300071, China
| | - Pengpeng Qu
- Department of Gynecological Oncology, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin 300071, China
| | - Zhaofeng Luo
- The Cancer Hospital of the University of Chinese Academy of Sciences, Aptamer Selection Center, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Liyun Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300350, China
| |
Collapse
|
29
|
A nucleolin-activated polyvalent aptamer nanoprobe for the detection of cancer cells. Anal Bioanal Chem 2023; 415:2217-2226. [PMID: 36864310 DOI: 10.1007/s00216-023-04629-3] [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: 01/07/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Abstract
Sensitive detection of cancer cells plays a critical role in early cancer diagnosis. Nucleolin, overexpressed on the surface of cancer cells, is regarded as a candidate biomarker for cancer diagnosis. Thus, cancer cells can be detected through the detection of membrane nucleolin. Herein, we designed a nucleolin-activated polyvalent aptamer nanoprobe (PAN) to detect cancer cells. In brief, a long single-stranded DNA with many repeated sequences was synthesized through rolling circle amplification (RCA). Then the RCA product acted as a scaffold chain to combine with multiple AS1411 sequences, which was doubly modified with fluorophore and quenching group, respectively. The fluorescence of PAN was initially quenched. Upon binding to target protein, the conformation of PAN changed, leading to the recovery of fluorescence. The fluorescence signal of cancer cells treated with PAN was much brighter compared with that of monovalent aptamer nanoprobes (MAN) at the same concentration. Furthermore, the binding affinity of PAN to B16 cells was proved to be 30 times higher than that of MAN by calculating the dissociation constants. The results indicated that PAN could specifically detect target cells, and this design concept has potential to become promising in cancer diagnosis.
Collapse
|
30
|
Jin B, Guo Z, Chen Z, Chen H, Li S, Deng Y, Jin L, Liu Y, Zhang Y, He N. Aptamers in cancer therapy: problems and new breakthroughs. J Mater Chem B 2023; 11:1609-1627. [PMID: 36744587 DOI: 10.1039/d2tb02579e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Aptamers, a class of oligonucleotides that can bind with molecular targets with high affinity and specificity, have been widely applied in research fields including biosensing, imaging, diagnosing, and therapy of diseases. However, compared with the rapid development in the research fields, the clinical application of aptamers is progressing at a much slower speed, especially in the therapy of cancer. Obstructions including nuclease degradation, renal clearance, a complex selection process, and potential side effects have inhibited the clinical transformation of aptamer-conjugated drugs. To overcome these problems, taking certain measures to improve the biocompatibility and stability of aptamer-conjugated drugs in vivo is necessary. In this review, the obstructions mentioned above are thoroughly discussed and the methods to overcome these problems are introduced in detail. Furthermore, landmark research works and the most recent studies on aptamer-conjugated drugs for cancer therapy are also listed as examples, and the future directions of research for aptamer clinical transformation are discussed.
Collapse
Affiliation(s)
- Baijiang Jin
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Zhukang Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Zhu Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Hui Chen
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Song Li
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Yan Deng
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Lian Jin
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Yuan Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yuanying Zhang
- Department of Molecular Biology, Jiangsu Cancer Hospital, Nanjing 210009, P. R. China
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China. .,Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| |
Collapse
|
31
|
Li P, Wang C, Wang W, Duan X, Li J. Preliminary evaluation of a 64Cu-labeled DNA aptamer for PET imaging of glioblastoma. J Radioanal Nucl Chem 2023. [DOI: 10.1007/s10967-023-08835-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
AbstractTo develop a DNA aptamer-based PET tracer for imaging of glioblastoma. 5 mM of NOTA-AS1411, 60-min, and 37 °C were selected as the optimal condition for 64Cu radiolabeling of AS1411. 64Cu-NOTA-AS1411 remained stable in PBS and 100% mouse serum for at least six hours. From the PET images, 64Cu-NOTA-AS1411 tended to be excreted out through the kidneys and there was high tracer accumulation in the bladder. There was a higher tumor uptake in the AS1411 group than that in the control group. 64Cu-NOTA-AS1411 is a suitable potential PET tracer for imaging murine glioblastoma.
Collapse
|
32
|
Zhan M, Guo Y, Shen M, Shi X. Nanomaterial‐Boosted Tumor Immunotherapy Through Natural Killer Cells. ADVANCED NANOBIOMED RESEARCH 2022; 2. [DOI: 10.1002/anbr.202200096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Natural killer (NK)‐cell immunotherapy as an alternative to T‐cell immunotherapy has been widely used in clinical cell immunotherapy of various tumors. Despite the surprising findings, the widespread applications of NK cells are still limited by the insufficient expansion and short lifespan of adoptive NK cells in vivo, the poor penetration of NK cells in solid tumors, as well as the immunosuppressive tumor microenvironment that may cause the inactivation of NK cells. Fortunately, the emergence of nanomaterials provides many opportunities to address these vexing problems, thus overcoming the barriers faced by NK cells and promoting the tumor inhibitory efficacy of NK cells. Herein, the recent advances in the rational design of nanomaterials for boosting the NK cell‐based immunotherapy, mainly through enhancing NK cell engagement with tumors, boosting NK cell activation or expansion, as well as redirecting NK cells to tumor cells, are reviewed. Lastly, the design and preparation of next‐generation nanomaterials that aim to further boost the NK cell‐based immunotherapy are briefly discussed.
Collapse
Affiliation(s)
- Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| |
Collapse
|
33
|
Fadeev M, O’Hagan MP, Biniuri Y, Willner I. Aptamer-Protein Structures Guide In Silico and Experimental Discovery of Aptamer-Short Peptide Recognition Complexes or Aptamer-Amino Acid Cluster Complexes. J Phys Chem B 2022; 126:8931-8939. [PMID: 36315022 PMCID: PMC9661473 DOI: 10.1021/acs.jpcb.2c05624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A method to computationally and experimentally identify aptamers against short peptides or amino acid clusters is introduced. The method involves the selection of a well-defined protein aptamer complex and the extraction of the peptide sequence participating in the binding of the protein to the aptamer. The subsequent fragmentation of the peptide sequence into short peptides and the in silico docking-guided identification of affinity complexes between the miniaturized peptides and the antiprotein aptamer, followed by experimental validation of the binding features of the short peptides with the antiprotein aptamers, leads to the identification of new short peptide-aptamer complexes. This is exemplified with the identification of the pentapeptide RYERN as the scaffold that binds thrombin to the DNA thrombin aptamer (DNA TA). In silico docking studies followed by microscale thermophoresis (MST) experiments demonstrate that the miniaturized tripeptides RYE, YER, and ERN reveal selective binding affinities toward the DNA TA. In addition, docking and MST experiments show that the ribonucleotide-translated RNA TA shows related binding affinities of YER to the DNA TA. Most importantly, we demonstrate that the separated amino acids Y/E/R assemble as a three amino acid cluster on the DNA TA and RNA TA aptamers in spatial configurations similar to the tripeptide YER on the respective aptamers. The clustering phenomenon is selective for the YER tripeptide system. The method to identify binding affinities of miniaturized peptides to known antiprotein aptamers and the specific clustering of single amino acids on the aptamers is further demonstrated by in silico and experimental identification of the binding of the tripeptide RET and the selective clustering of the separated amino acids R/E/T onto a derivative of the AS1411 aptamer against the nucleolin receptor protein.
Collapse
|
34
|
Li D, Ling S, Meng D, Zhou B, Liang P, Lv B. Sensitive fluorescent aptasensing of tobramycin on graphene oxide coupling strand displacement amplification and hybridization chain reaction. Int J Biol Macromol 2022; 220:1287-1293. [PMID: 36037911 DOI: 10.1016/j.ijbiomac.2022.08.158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022]
Abstract
An ultrasensitive biosensor was designed and constructed for tobramycin detection. As a target recognition component, the DNA probe consists of an aptamer region for tobramycin binding and a template for amplification. In the absence of tobramycin, the probe was locked to form a stem-loop structure. In the presence of the target, the binding of tobramycin led to a conformational change in the probe. The released 3' end was used as a primer for the strand displacement amplification (SDA) to produce a large amount of single-stranded trigger DNA, which then efficiently initiated the following hybridization chain reaction (HCR) to produce a long duplex DNA with many fluorophores. The signals were detected after the addition of graphene oxide (GO) to quench the fluorescence from excess hairpin DNA. Through sequence and reaction condition optimization, the biosensor exhibited high selectivity for tobramycin. The linearity range and limit of detection (LOD) were 0.5-30 nM and 0.06 nM, respectively. Moreover, the application of detecting tobramycin in milk and lake water samples showed that this method is reliable and could be further used in food safety control and environmental monitoring.
Collapse
Affiliation(s)
- Dawei Li
- Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China.
| | - Shen Ling
- Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China
| | - Dudu Meng
- Jiangsu Key Laboratory for Biofunctional Molecules, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China
| | - Bing Zhou
- Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China
| | - Pengda Liang
- Co-Innovation Center for Sustainable Forestry in Southern China, Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Biology and the Environment, Nanjing Forestry University, Nanjing 210037, China
| | - Bei Lv
- Jiangsu Key Laboratory for Biofunctional Molecules, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China.
| |
Collapse
|
35
|
Wang L, Lan H, Guan W, Han J, Liu Y, Wang Y, Mao Y, Wang Y. One-step Purification of Target Enzymes Using Interaction- and Structure-Based Design of Aptamer-Affinity Responsive Polymers: Selective Immobilization and Enhanced Stability. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2022.122758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Chen J, Xu J, Xiang J, Wan T, Deng H, Li D. A multivalent activatable aptamer probe with ultralow background signal and high sensitivity for diagnosis of lung adenocarcinoma. Talanta 2022. [DOI: 10.1016/j.talanta.2022.124056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
37
|
Abstract
The homeostasis of cellular activities is essential for the normal functioning of living organisms. Hence, the ability to regulate the fates of cells is of great significance for both fundamental chemical biology studies and therapeutic development. Despite the notable success of small-molecule drugs that normally act on cellular protein functions, current clinical challenges have highlighted the use of macromolecules to tune cell function for improved therapeutic outcomes. As a class of hybrid biomacromolecules gaining rapidly increasing attention, protein conjugates have exhibited great potential as versatile tools to manipulate cell function for therapeutic applications, including cancer treatment, tissue engineering, and regenerative medicine. Therefore, recent progress in the design and assembly of protein conjugates used to regulate cell function is discussed in this review. The protein conjugates covered here are classified into three different categories based on their mechanisms of action and relevant applications: (1) regulation of intercellular interactions; (2) intervention in intracellular biological pathways; (3) termination of cell proliferation. Within each genre, a variety of protein conjugate scaffolds are discussed, which contain a diverse array of grafted molecules, such as lipids, oligonucleotides, synthetic polymers, and small molecules, with an emphasis on their conjugation methodologies and potential biomedical applications. While the current generation of protein conjugates is focused largely on delivery, the next generation is expected to address issues of site-specific conjugation, in vivo stability, controllability, target selectivity, and biocompatibility.
Collapse
Affiliation(s)
- Yiao Wang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
38
|
Chen X, Ma Y, Xie Y, Pu J. Aptamer-based applications for cardiovascular disease. Front Bioeng Biotechnol 2022; 10:1002285. [PMID: 36312558 PMCID: PMC9606242 DOI: 10.3389/fbioe.2022.1002285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease (especially atherosclerosis) is a major cause of death worldwide, and novel diagnostic tools and treatments for this disease are urgently needed. Aptamers are single-stranded oligonucleotides that specifically recognize and bind to the targets by forming unique structures in vivo, enabling them to rival antibodies in cardiac applications. Chemically synthesized aptamers can be readily modified in a site-specific way, so they have been engineered in the diagnosis of cardiac diseases and anti-thrombosis therapeutics. Von Willebrand Factor plays a unique role in the formation of thrombus, and as an aptamer targeting molecule, has shown initial success in antithrombotic treatment. A combination of von Willebrand Factor and nucleic acid aptamers can effectively inhibit the progression of blood clots, presenting a positive diagnosis and therapeutic effect, as well as laying a novel theory and strategy to improve biocompatibility paclitaxel drug balloon or implanted stent in the future. This review summarizes aptamer-based applications in cardiovascular disease, including biomarker discovery and future management strategy. Although relevant applications are relatively new, the significant advancements achieved have demonstrated that aptamers can be promising agents to realize the integration of diagnosis and therapy in cardiac research.
Collapse
Affiliation(s)
| | | | | | - Jun Pu
- *Correspondence: Yuquan Xie, ; Jun Pu,
| |
Collapse
|
39
|
Li R, Wu X, Li J, Lu X, Zhao RC, Liu J, Ding B. A covalently conjugated branched DNA aptamer cluster-based nanoplatform for efficiently targeted drug delivery. NANOSCALE 2022; 14:9369-9378. [PMID: 35726974 DOI: 10.1039/d2nr01252a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Targeted delivery of therapeutic drugs is essential for precise treatment of various diseases to reduce possible serious side-effects. A screened DNA aptamer has been widely developed for active targeting delivery. Herein, we report a facile strategy for the construction of a branched DNA aptamer cluster-based nanoplatform for efficiently targeted drug delivery. In our design, the terminal-modified DNA aptamer can be covalently conjugated to form a branched aptamer cluster by click reaction easily. The branched aptamer cluster-modified DNA tetrahedron (TET) demonstrates highly targeted cellular uptake with the modification of only one site. After loading the chemotherapeutic drug (doxorubicin, DOX), the DNA aptamer cluster-based nanoplatform elicits a remarkable and selective inhibition of tumor cell proliferation by much-enhanced targeted delivery. This covalently conjugated branched DNA aptamer cluster-based nanoplatform provides a new strategy for the development of targeted drug delivery.
Collapse
Affiliation(s)
- Runze Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Dongdan Santiao No. 5, Beijing 100005, China.
| | - Xiaohui Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Dongdan Santiao No. 5, Beijing 100005, China.
| | - Xuehe Lu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Dongdan Santiao No. 5, Beijing 100005, China.
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jianbing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, 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, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
40
|
Aptamers as Recognition Elements for Electrochemical Detection of Exosomes. Chem Res Chin Univ 2022; 38:879-885. [PMID: 35578711 PMCID: PMC9094132 DOI: 10.1007/s40242-022-2088-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/11/2022] [Indexed: 11/08/2022]
Abstract
Exosome analysis is emerging as an attractive noninvasive approach for disease diagnosis and treatment monitoring in the field of liquid biopsy. Aptamer is considered as a promising molecular probe for exosomes detection because of the high binding affinity, remarkable specificity, and low cost. Recently, many approaches have been developed to further improve the performance of electrochemical aptamer based(E-AB) sensors with a lower limit of detection. In this review, we focus on the development of using aptamer as a specific recognition element for exosomes detection in electrochemical sensors. We first introduce recent advances in evolving aptamers against exosomes. Then, we review methods of immobilization aptamers on electrode surfaces, followed by a summary of the main strategies of signal amplification. Finally, we present the insights of the challenges and future directions of E-AB sensors for exosomes analysis.
Collapse
|
41
|
Chen S, Zhang L, Yuan Q, Tan J. Current Advances in Aptamer-based Biomolecular Recognition and Biological Process Regulation. Chem Res Chin Univ 2022; 38:847-855. [PMID: 35573821 PMCID: PMC9077342 DOI: 10.1007/s40242-022-2087-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/08/2022] [Indexed: 12/01/2022]
Abstract
The interaction between biomolecules with their target ligands plays a great role in regulating biological functions. Aptamers are short oligonucleotide sequences that can specifically recognize target biomolecules via structural complementarity and thus regulate related biological functions. In the past ten years, aptamers have made great progress in target biomolecule recognition, becoming a powerful tool to regulate biological functions. At present, there are many reviews on aptamers applied in biomolecular recognition, but few reviews pay attention to aptamer-based regulation of biological functions. Here, we summarize the approaches to enhancing aptamer affinity and the advancements of aptamers in regulating enzymatic activity, cellular immunity and cellular behaviors. Furthermore, this review discusses the challenges and future perspectives of aptamers in target recognition and biological functions regulation, aiming to provide some promising ideas for future regulation of biomolecular functions in a complex biological environment.
Collapse
Affiliation(s)
- Sisi Chen
- Molecular Science and Biomedicine Laboratory(MBL), Institute of Chemical Biology and Nanomedicine(ICBN), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082 P. R. China
| | - Lei Zhang
- Molecular Science and Biomedicine Laboratory(MBL), Institute of Chemical Biology and Nanomedicine(ICBN), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082 P. R. China
| | - Quan Yuan
- Molecular Science and Biomedicine Laboratory(MBL), Institute of Chemical Biology and Nanomedicine(ICBN), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082 P. R. China
| | - Jie Tan
- Molecular Science and Biomedicine Laboratory(MBL), Institute of Chemical Biology and Nanomedicine(ICBN), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082 P. R. China
| |
Collapse
|
42
|
Xu J, Xiang J, Chen J, Wan T, Deng H, Li D. High sensitivity detection of tumor cells in biological samples using a multivalent aptamer strand displacement strategy. Analyst 2022; 147:634-644. [PMID: 35040831 DOI: 10.1039/d1an01949j] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Monitoring the cell surface-expressed nucleolin facilitates early cancer diagnosis. Herein, we developed a multivalent aptamer displacement strand duplex strategy on cell membranes using a multi-receptor co-recognition design for improving the sensitivity and specificity of cancer cell recognition with an ultra-low background. The AS1411 aptamer labeled with the FAM fluorophore can be quenched using a partial complementary sequence modified with a BHQ1 tag which is partially hybridized with the AS1411 aptamer to create a receptor-activating aptamer. The multi-AS1411 activable probe based on the strand displacement strategy was constructed using multiple copies of the structure-switching AS1411 aptamer (bearing a short poly-A tail) linked together using the poly-T long chain (as a scaffold) which was synthesized by Terminal Deoxynucleotidyl Transferase (TDT)-mediated extension. We demonstrated the promising efficacy and sensitivity of our method in recognizing tumor cells in both cell mixtures and clinical cytology specimens. Due to its simple and fast operation with excellent cell recognition sensitivity and accuracy, it is expected to achieve the detection of low abundance target cells. Our approach will have broad application in clinical rapid detection and personalized medicine.
Collapse
Affiliation(s)
- Jieru Xu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Jiahui Xiang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Jialing Chen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Tao Wan
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Hongli Deng
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Dairong Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
43
|
Qi S, Duan N, Khan IM, Dong X, Zhang Y, Wu S, Wang Z. Strategies to manipulate the performance of aptamers in SELEX, post-SELEX and microenvironment. Biotechnol Adv 2022; 55:107902. [DOI: 10.1016/j.biotechadv.2021.107902] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023]
|
44
|
Haberland A, Müller J. Aptamers Against COVID-19: An Untested Opportunity. Mini Rev Med Chem 2022; 22:1708-1715. [PMID: 35023454 PMCID: PMC9896377 DOI: 10.2174/1389557522666220112094951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/22/2021] [Accepted: 11/08/2021] [Indexed: 11/22/2022]
Abstract
Given the lack of success in the development of effective drugs to treat COVID-19, which show "game-changing" potential, it is necessary to explore drugs with different modes of action. Single mode-of-action drugs have not been succeeded in curing COVID-19, which is a highly complex disease. This is the case for direct antivirals and anti-inflammatory drugs, both of which treat different phases of the disease. Aptamers are molecules that deliver different modes of action, allowing their effects to be bundled, which, when combined, support their therapeutic efficacy. In this minireview, we summarise the current activities in the development of aptamers for the treatment of COVID-19 and long-COVID. A special emphasis is placed on the capability of their multiple modes of action, which is a promising approach for treating complex diseases such as COVID-19.
Collapse
Affiliation(s)
- Annekathrin Haberland
- Berlin Cures GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;,Address correspondence to this author at the Berlin Cures GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; E-mail:
| | - Johannes Müller
- Berlin Cures GmbH, Knesebeckstr. 59-61, 10719 Berlin, Germany
| |
Collapse
|
45
|
Zhang Z, Pandey R, Li J, Gu J, White D, Stacey HD, Ang JC, Steinberg C, Capretta A, Filipe CDM, Mossman K, Balion C, Miller MS, Salena BJ, Yamamura D, Soleymani L, Brennan JD, Li Y. High‐Affinity Dimeric Aptamers Enable the Rapid Electrochemical Detection of Wild‐Type and B.1.1.7 SARS‐CoV‐2 in Unprocessed Saliva. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202110819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Zijie Zhang
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
| | - Richa Pandey
- Department of Engineering Physics McMaster University Canada
| | - Jiuxing Li
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
| | - Dawn White
- Biointerfaces Institute McMaster University Canada
| | - Hannah D. Stacey
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- McMaster Immunology Research Centre McMaster University Canada
| | - Jann C. Ang
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- McMaster Immunology Research Centre McMaster University Canada
| | | | - Alfredo Capretta
- Biointerfaces Institute McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
| | | | - Karen Mossman
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- Department of Medicine McMaster University Canada
| | - Cynthia Balion
- Department of Pathology and Molecular Medicine McMaster University Canada
| | - Matthew S. Miller
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- McMaster Immunology Research Centre McMaster University Canada
| | | | - Deborah Yamamura
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- Department of Pathology and Molecular Medicine McMaster University Canada
| | - Leyla Soleymani
- Department of Engineering Physics McMaster University Canada
- School of Biomedical Engineering McMaster University 1280 Main Street West Hamilton Ontario L8S 4K1 Canada
| | | | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences McMaster University Canada
- Biointerfaces Institute McMaster University Canada
- Michael G. DeGroote Institute of Infectious Disease Research McMaster University Canada
- School of Biomedical Engineering McMaster University 1280 Main Street West Hamilton Ontario L8S 4K1 Canada
| |
Collapse
|
46
|
Zhang Z, Pandey R, Li J, Gu J, White D, Stacey HD, Ang JC, Steinberg C, Capretta A, Filipe CDM, Mossman K, Balion C, Miller MS, Salena BJ, Yamamura D, Soleymani L, Brennan JD, Li Y. High-Affinity Dimeric Aptamers Enable the Rapid Electrochemical Detection of Wild-Type and B.1.1.7 SARS-CoV-2 in Unprocessed Saliva. Angew Chem Int Ed Engl 2021; 60:24266-24274. [PMID: 34464491 PMCID: PMC8596624 DOI: 10.1002/anie.202110819] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Indexed: 01/05/2023]
Abstract
We report a simple and rapid saliva-based SARS-CoV-2 antigen test that utilizes a newly developed dimeric DNA aptamer, denoted as DSA1N5, that specifically recognizes the spike proteins of the wildtype virus and its Alpha and Delta variants with dissociation constants of 120, 290 and 480 pM, respectively, and binds pseudotyped lentiviruses expressing the wildtype and alpha trimeric spike proteins with affinity constants of 2.1 pM and 2.3 pM, respectively. To develop a highly sensitive test, DSA1N5 was immobilized onto gold electrodes to produce an electrochemical impedance sensor, which was capable of detecting 1000 viral particles per mL in 1:1 diluted saliva in under 10 min without any further sample processing. Evaluation of 36 positive and 37 negative patient saliva samples produced a clinical sensitivity of 80.5 % and specificity of 100 % and the sensor could detect the wildtype virus as well as the Alpha and Delta variants in the patient samples, which is the first reported rapid test that can detect any emerging variant of SARS-CoV-2.
Collapse
Affiliation(s)
- Zijie Zhang
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
| | - Richa Pandey
- Department of Engineering PhysicsMcMaster UniversityCanada
| | - Jiuxing Li
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
| | - Dawn White
- Biointerfaces InstituteMcMaster UniversityCanada
| | - Hannah D. Stacey
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- McMaster Immunology Research CentreMcMaster UniversityCanada
| | - Jann C. Ang
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- McMaster Immunology Research CentreMcMaster UniversityCanada
| | | | - Alfredo Capretta
- Biointerfaces InstituteMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
| | | | - Karen Mossman
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- Department of MedicineMcMaster UniversityCanada
| | - Cynthia Balion
- Department of Pathology and Molecular MedicineMcMaster UniversityCanada
| | - Matthew S. Miller
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- McMaster Immunology Research CentreMcMaster UniversityCanada
| | | | - Deborah Yamamura
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- Department of Pathology and Molecular MedicineMcMaster UniversityCanada
| | - Leyla Soleymani
- Department of Engineering PhysicsMcMaster UniversityCanada
- School of Biomedical EngineeringMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | | | - Yingfu Li
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityCanada
- Biointerfaces InstituteMcMaster UniversityCanada
- Michael G. DeGroote Institute of Infectious Disease ResearchMcMaster UniversityCanada
- School of Biomedical EngineeringMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| |
Collapse
|
47
|
Lv Z, Wang Q, Yang M. Multivalent Duplexed-Aptamer Networks Regulated a CRISPR-Cas12a System for Circulating Tumor Cell Detection. Anal Chem 2021; 93:12921-12929. [PMID: 34533940 DOI: 10.1021/acs.analchem.1c02228] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although circulating tumor cells (CTCs) have great potential to act as the mini-invasive liquid biopsy cancer biomarker, a rapid and sensitive CTC detection method remains lacking. CRISPR-Cas12a has recently emerged as a promising tool in biosensing applications with the characteristic of fast detection, easy operation, and high sensitivity. Herein, we reported a CRISPR-Cas12a-based CTC detection sensor that is regulated by the multivalent duplexed-aptamer networks (MDANs). MDANs were synthesized on a magnetic bead surface by rolling circle amplification (RCA), which contain multiple duplexed-aptamer units that allow structure switching induced by cell-binding events. The presence of target cells can trigger the release of free "activator DNA" from the MDANs structure to activate the downstream CRISPR-Cas12a for signal amplification. Furthermore, the 3D DNA network formed by RCA products also provided significantly higher sensitivity than the monovalent aptamer. As a proof-of-concept study, we chose the most widely used sgc8 aptamer that specifically recognizes CCRF-CEM cells to validate the proposed approach. The MDANs-Cas12a system could afford a simple and fast CTC detection workflow with a detection limit of 26 cells mL-1. We also demonstrated that the MDANs-Cas12a could directly detect the CTCs in human blood samples, indicating a great potential of the MDANs-Cas12a in clinical CTC-based liquid biopsy.
Collapse
Affiliation(s)
- Zhengxian Lv
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering and State Key Lab of Marine Environmental Science, Xiamen University, Xiamen 361005, China
| | - Qiuquan Wang
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering and State Key Lab of Marine Environmental Science, Xiamen University, Xiamen 361005, China
| | - Minghui Yang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
48
|
Yan J, Gao T, Lu Z, Yin J, Zhang Y, Pei R. Aptamer-Targeted Photodynamic Platforms for Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:27749-27773. [PMID: 34110790 DOI: 10.1021/acsami.1c06818] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Achieving controlled and accurate delivery of photosensitizers (PSs) into tumor sites is a major challenge in conventional photodynamic therapy (PDT). Aptamer is a short oligonucleotide sequence (DNA or RNA) with a folded three-dimensional structure, which can selectively bind to specific small molecules, proteins, or the whole cells. Aptamers could act as ligands and be modified onto PSs or nanocarriers, enabling specific recognition and binding to tumor cells or their membrane proteins. The resultant aptamer-modified PSs or PSs-containing nanocarriers generate amounts of reactive oxygen species with light irradiation and obtain superior photodynamic therapeutic efficiency in tumors. Herein, we overview the recent progress in the designs and applications of aptamer-targeted photodynamic platforms for tumor therapy. First, we focus on the progress on the rational selection of aptamers and summarize the applications of aptamers which have been applied for targeted tumor diagnosis and therapy. Then, aptamer-targeted photodynamic therapies including various aptamer-PSs, aptamer-nanocarriers containing PSs, and aptamer-nano-photosensitizers are highlighted. The aptamer-targeted synergistically therapeutic platforms including PDT, photothermal therapy, and chemotherapy, as well as the imaging-guided theranostics, are also discussed. Finally, we offer an insight into the development trends and future perspectives of aptamer-targeted photodynamic platforms for tumor therapy.
Collapse
Affiliation(s)
- Jincong Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Tian Gao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Zhongzhong Lu
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 200444 Shanghai, China
| | - Ye Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, 215123 Suzhou, China
| |
Collapse
|
49
|
Shi X, Chen L, Chen S, Sun D. Electrochemical aptasensors for the detection of hepatocellular carcinoma-related biomarkers. NEW J CHEM 2021. [DOI: 10.1039/d1nj01042e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent progress in electrochemical aptasensors for the detection of HCC-related biomarkers, including cancer cells, proteins, cell-derived exosomes, and nucleic acids, is reviewed.
Collapse
Affiliation(s)
- Xianhua Shi
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Linxi Chen
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Siyi Chen
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Duanping Sun
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Laboratory of New Drug Discovery and Evaluation of Ordinary Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| |
Collapse
|