1
|
Chatterjee O, Kaur GA, Shukla N, Balayan S, Singh PK, Chatterjee S, Tiwari A. Multifaceted arsenal in SELEX nanomedicine. Adv Colloid Interface Sci 2025; 342:103540. [PMID: 40344950 DOI: 10.1016/j.cis.2025.103540] [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: 02/06/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Aptamers, short oligonucleotide sequences that bind specifically to cellular proteins and receptors, are emerging as versatile tools in molecular nanomedicine. Unlike passive tumor targeting via the enhanced permeability and retention (EPR) effect, aptamers enable precise drug delivery, enhancing therapeutic efficacy while minimizing side effects. Developed through the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process, aptamers offer compact size, robust structure, chemical versatility, and cost-effective synthesis. They serve as effective delivery vehicles for therapeutic molecules, including miRNA, siRNA, and small-molecule drugs, and function as antibody-like ligands for applications in cancer, diabetes, and autoimmune disorders. Since the approval of Macugen, the first aptamer targeting VEGF, aptamers have also shown promise as diagnostic sensors and theranostic agents. This review explores SELEX-derived aptamers in nanomedicine, focusing on their therapeutic and diagnostic roles, particularly in precision cancer therapies. It also addresses challenges such as degradation and clinical translation alongside prospects in vaccines, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Oishika Chatterjee
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden; Department of Biological Sciences, Bose Institute Unified Academic Campus EN 80, Sector 5, Bidhan Nagar (Salt Lake City) Kolkata 700 091, WB, India
| | - Gun Anit Kaur
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Nutan Shukla
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Sapna Balayan
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Pravin Kumar Singh
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Subhrangsu Chatterjee
- Department of Biological Sciences, Bose Institute Unified Academic Campus EN 80, Sector 5, Bidhan Nagar (Salt Lake City) Kolkata 700 091, WB, India.
| | - Ashutosh Tiwari
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden.
| |
Collapse
|
2
|
Ning Y, Li B, Chen W, Feng L, Huang X, Liu B. DNA Framework-Based Lysosome-Targeting Chimeras: Intracellular ATP-Facilitated Extracellular Protein Degradation. ACS NANO 2025; 19:15853-15862. [PMID: 40237339 DOI: 10.1021/acsnano.5c00745] [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: 04/18/2025]
Abstract
Targeted protein degradation (TPD) offered a riveting therapeutic paradigm to eradicate pathogenesis-relevant proteins, especially those belonging to the once-considered undruggable proteome. Considering that adenosine triphosphate (ATP) is the primary energy source for cell activities and lysosomes are important ATP storage sites, herein, the first example of dual-function tetrahedral DNA framework-based lysosome-targeting chimeras (TDF-LYTACs) is proposed for elucidating the correlation between extracellular protein degradation via the lysosome pathway and the fluctuations in intracellular ATP levels. In our study, platelet-derived growth factor (PDGF), a driver of cancer invasion and metastasis, was chosen as the protein of interest. To achieve multifunctionality, we employed a tetrahedral DNA framework formed by an aptamer of PDGF, human apurinic/apyrimidinic endonuclease 1 (APE1)-triggered ATP probes, and a ligand of the cell-surface lysosome-shuttling receptor (IGFIIR). TDF-LYTACs efficiently and quickly shuttled PDGF proteins to lysosomes, degraded them through the lysosomal pathway, and further visualized the intracellular ATP level synchronously. Furthermore, we found a significant correlation between the degradation efficiency of PDGF and intracellular ATP levels over time; that is, a higher ATP level corresponded to higher degradation efficiency and vice versa. We anticipate that our versatile TDF-LYTACs will offer a perspective for degrading multifunctional extracellular proteins.
Collapse
Affiliation(s)
- Yujun Ning
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Bin Li
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Weishuai Chen
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Langxia Feng
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Xuedong Huang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers and Institute of Biomedical Sciences, Fudan University, Shanghai 200433, People's Republic of China
| |
Collapse
|
3
|
Huang C, Harris KS, Siddiqui G, Jörg M. Recommended Tool Compounds: Thienotriazolodiazepines-Derivatized Chemical Probes to Target BET Bromodomains. ACS Pharmacol Transl Sci 2025; 8:978-1012. [PMID: 40242580 PMCID: PMC11997894 DOI: 10.1021/acsptsci.4c00726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/29/2025] [Accepted: 02/18/2025] [Indexed: 04/18/2025]
Abstract
Thienotriazolodiazepines, including (+)-JQ1 (4), are well-known inhibitors of the bromodomain (BD) and extra-terminal domain (BET) family of proteins. Despite the suboptimal physicochemical properties as a drug candidate, such as poor solubility and half-life, (+)-JQ1 (4) has proven as an effective chemical probe with high target potency and selectivity. (+)-JQ1 (4) and (+)-JQ1-derived chemical probes have played a vital role in chemical biology and drug discovery over the past decade, which is demonstrated by the high number of impactful research studies published since the disclosure of (+)-JQ1 (4) in 2010. In this review, we discuss the development of (+)-JQ1-derivatized chemical probes over the past decade and their significant contribution to scientific research. Specifically, we will summarize the development of innovative label-free and labeled (+)-JQ1-derivatized chemical probes, such as bivalent, covalent, and photoaffinity probes as well as protein degraders, with a focus on the design of these chemical probes.
Collapse
Affiliation(s)
- Chuhui Huang
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- Drug
Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical
Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Kate S. Harris
- Chemistry-School
of Natural and Environmental Sciences, Newcastle
University, Newcastle
Upon Tyne NE1 7RU, United Kingdom
| | - Ghizal Siddiqui
- Drug
Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical
Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Manuela Jörg
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- Chemistry-School
of Natural and Environmental Sciences, Newcastle
University, Newcastle
Upon Tyne NE1 7RU, United Kingdom
| |
Collapse
|
4
|
Hsu YW, Ma L, Tang Y, Li M, Zhou C, Geng Y, Zhang C, Wang T, Guo W, Li M, Wang Y. The application of aptamers in the repair of bone, nerve, and vascular tissues. J Mater Chem B 2025; 13:1872-1889. [PMID: 39760465 DOI: 10.1039/d4tb02180k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Aptamers represent a distinct category of short nucleotide sequences or peptide molecules characterized by their ability to bind to specific targets with high precision. These molecules are predominantly synthesized through SELEX (Systematic Evolution of Ligands by Exponential Enrichment) technology. Recent findings indicate that aptamers may have significant applications in regenerative medicine, particularly in the domain of tissue repair. In comparison to other bioactive agents, aptamers exhibit superior specificity and affinity, are more readily accessible, and can be chemically modified, thereby presenting a promising avenue for the functionalization of tissue engineering materials in tissue repair applications. This review delineates the properties of aptamers and examines the methodologies and advancements related to aptamer-functionalized hydrogels, nanoparticles, and electrospun materials. It categorizes the four primary functions of aptamers in tissue repair, namely regeneration, delivery systems, anti-inflammatory actions, and pro-coagulation effects. Furthermore, the review explores the utilization of aptamer-functionalized tissue engineering materials in the repair of bone, nerve, and vascular tissues, highlighting the mechanisms by which aptamers facilitate tissue growth and repair through regenerative properties and their role in transporting substances that promote repair. Lastly, the review addresses the future prospects and challenges associated with the application of aptamers in tissue repair, offering novel insights and directions for further research and application in this domain.
Collapse
Affiliation(s)
- Yu-Wei Hsu
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Emergency Department, Peking University People's Hospital, Beijing, 100044, China.
| | - Le Ma
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Ye Tang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, 100044, China
| | - Mengen Li
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, 100044, China
| | - Chengkai Zhou
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Yan Geng
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Chenxi Zhang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Tianbing Wang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Wei Guo
- Emergency Department, Peking University People's Hospital, Beijing, 100044, China.
| | - Ming Li
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Yanhua Wang
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
5
|
Hu J, Gao D. Recent Advances in Aptamer-Based Microfluidic Biosensors for the Isolation, Signal Amplification and Detection of Exosomes. SENSORS (BASEL, SWITZERLAND) 2025; 25:848. [PMID: 39943486 PMCID: PMC11820184 DOI: 10.3390/s25030848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025]
Abstract
Exosomes carry diverse tumor-associated molecular information that can reflect real-time tumor progression, making them a promising tool for liquid biopsy. However, traditional methods for exosome isolation and detection often rely on large, expensive equipment and are time-consuming, limiting their practical applicability in clinical settings. Microfluidic technology offers a versatile platform for exosome analysis, with advantages such as seamless integration, portability and reduced sample volumes. Aptamers, which are single-stranded oligonucleotides with high affinity and specificity for target molecules, have been frequently employed in the development of aptamer-based microfluidics for the isolation, signal amplification, and quantitative detection of exosomes. This review summarizes recent advances in aptamer-based microfluidic strategies for exosome analysis, including (1) strategies for on-chip exosome capture mediated by aptamers combined with nanomaterials or nanointerfaces; (2) aptamer-based on-chip signal amplification techniques, such as enzyme-free hybridization chain reaction (HCR), rolling circle amplification (RCA), and DNA machine-assisted amplification; and (3) various aptamer-assisted detection methods, such as fluorescence, electrochemistry, surface-enhanced Raman scattering (SERS), and magnetism. The limitations and advantages of these methods are also summarized. Finally, future challenges and directions for the clinical analysis of exosomes based on aptamer-based microfluidics are discussed.
Collapse
Affiliation(s)
- Jessica Hu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School and Open FIESTA, Tsinghua University, Shenzhen 518055, China;
- Key Laboratory of Metabolomics at Shenzhen, Shenzhen 518055, China
| | - Dan Gao
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School and Open FIESTA, Tsinghua University, Shenzhen 518055, China;
- Key Laboratory of Metabolomics at Shenzhen, Shenzhen 518055, China
| |
Collapse
|
6
|
Yim J, Park J, Kim G, Lee HH, Chung JS, Jo A, Koh M, Park J. Conditional PROTAC: Recent Strategies for Modulating Targeted Protein Degradation. ChemMedChem 2024; 19:e202400326. [PMID: 38993102 PMCID: PMC11581424 DOI: 10.1002/cmdc.202400326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) have emerged as a promising technology for inducing targeted protein degradation by leveraging the intrinsic ubiquitin-proteasome system (UPS). While the potential druggability of PROTACs toward undruggable proteins has accelerated their rapid development and the wide-range of applications across diverse disease contexts, off-tissue effects and side-effects of PROTACs have recently received attentions to improve their efficacy. To address these issues, spatial or temporal target protein degradation by PROTACs has been spotlighted. In this review, we explore chemical strategies for modulating protein degradation in a cell type-specific (spatio-) and time-specific (temporal-) manner, thereby offering insights for expanding PROTAC applications to overcome the current limitations of target protein degradation strategy.
Collapse
Affiliation(s)
- Junhyeong Yim
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Multidimensional Genomics Research CenterKangwon National UniversityChuncheon24341Republic of Korea
| | - Junyoung Park
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Institute for Molecular Science and Fusion TechnologyKangwon National UniversityChuncheon24341Republic of Korea
| | - Gabin Kim
- Department of Chemistry and ChemistryInstitute for Functional MaterialsPusan National UniversityBusan46241Republic of Korea
| | - Hyung Ho Lee
- Department of UrologyUrological Cancer CenterResearch Institute and Hospital of National Cancer CenterGoyang10408Republic of Korea
| | - Jin Soo Chung
- Department of UrologyUrological Cancer CenterResearch Institute and Hospital of National Cancer CenterGoyang10408Republic of Korea
| | - Ala Jo
- Center for NanomedicineInstitute for Basic ScienceSeoul03722Republic of Korea
| | - Minseob Koh
- Department of Chemistry and ChemistryInstitute for Functional MaterialsPusan National UniversityBusan46241Republic of Korea
| | - Jongmin Park
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Multidimensional Genomics Research CenterKangwon National UniversityChuncheon24341Republic of Korea
- Institute for Molecular Science and Fusion TechnologyKangwon National UniversityChuncheon24341Republic of Korea
| |
Collapse
|
7
|
Fàbrega C, Gallisà-Suñé N, Zuin A, Ruíz JS, Coll-Martínez B, Fabriàs G, Eritja R, Crosas B. Aptamer-Hytac Chimeras for Targeted Degradation of SARS-CoV-2 Spike-1. Cells 2024; 13:1767. [PMID: 39513874 PMCID: PMC11544835 DOI: 10.3390/cells13211767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
The development of novel tools to tackle viral processes has become a central focus in global health, during the COVID-19 pandemic. The spike protein is currently one of the main SARS-CoV-2 targets, owing to its key roles in infectivity and virion formation. In this context, exploring innovative strategies to block the activity of essential factors of SARS-CoV-2, such as spike proteins, will strengthen the capacity to respond to current and future threats. In the present work, we developed and tested novel bispecific molecules that encompass: (i) oligonucleotide aptamers S901 and S702, which bind to the spike protein through its S1 domain, and (ii) hydrophobic tags, such as adamantane and tert-butyl-carbamate-based ligands. Hydrophobic tags have the capacity to trigger the degradation of targets recruited in the context of a proteolytic chimera by activating quality control pathways. We observed that S901-adamantyl conjugates promote the degradation of the S1 spike domain, stably expressed in human cells by genomic insertion. These results highlight the suitability of aptamers as target-recognition molecules and the robustness of protein quality control pathways triggered by hydrophobic signals, and place aptamer-Hytacs as promising tools for counteracting coronavirus progression in human cells.
Collapse
Affiliation(s)
- Carme Fàbrega
- Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Núria Gallisà-Suñé
- Proteasome Regulation Lab, Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 4, 08028 Barcelona, Spain (A.Z.)
| | - Alice Zuin
- Proteasome Regulation Lab, Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 4, 08028 Barcelona, Spain (A.Z.)
| | - Juan Sebastián Ruíz
- Lincbiotech SL, Avenida do Mestre Mateo, 2, 15706 Santiago de Compostela, Spain;
| | - Bernat Coll-Martínez
- Proteasome Regulation Lab, Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 4, 08028 Barcelona, Spain (A.Z.)
| | - Gemma Fabriàs
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034 Barcelona, Spain;
| | - Ramon Eritja
- Department of Surfactants and Nanobiotechnology, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Bernat Crosas
- Proteasome Regulation Lab, Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 4, 08028 Barcelona, Spain (A.Z.)
| |
Collapse
|
8
|
Daneva GN, Tsiakanikas P, Adamopoulos PG, Scorilas A. Kallikrein-related peptidases: mechanistic understanding for potential therapeutic targeting in cancer. Expert Opin Ther Targets 2024; 28:875-894. [PMID: 39431595 DOI: 10.1080/14728222.2024.2415014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Human kallikrein-related peptidases (KLKs) represent a subgroup of 15 serine endopeptidases involved in various physiological processes and pathologies, including cancer. AREAS COVERED This review aims to provide a comprehensive overview of the KLK family, highlighting their genomic structure, expression profiles and substrate specificity. We explore the role of KLKs in tumorigenesis, emphasizing their potential as biomarkers and therapeutic targets in cancer treatment. The dysregulated activity of KLKs has been linked to various malignancies, making them promising candidates for cancer diagnostics and therapy. EXPERT OPINION : Recent advancements in understanding the mechanistic pathways of KLK-related tumorigenesis offer new prospects for developing targeted cancer treatments. Expert opinion suggests that while significant progress has been made, further research is necessary to fully exploit KLKs' potential in clinical applications.
Collapse
Affiliation(s)
- Glykeria N Daneva
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Tsiakanikas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
9
|
Kong L, Meng F, Zhou P, Ge R, Geng X, Yang Z, Li G, Zhang L, Wang J, Ma J, Dong C, Zhou J, Wu S, Zhong D, Xie S. An engineered DNA aptamer-based PROTAC for precise therapy of p53-R175H hotspot mutant-driven cancer. Sci Bull (Beijing) 2024; 69:2122-2135. [PMID: 38811338 DOI: 10.1016/j.scib.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/02/2024] [Accepted: 03/07/2024] [Indexed: 05/31/2024]
Abstract
Targeting oncogenic mutant p53 represents an attractive strategy for cancer treatment due to the high frequency of gain-of-function mutations and ectopic expression in various cancer types. Despite extensive efforts, the absence of a druggable active site for small molecules has rendered these mutants therapeutically non-actionable. Here we develop a selective and effective proteolysis-targeting chimera (PROTAC) for p53-R175H, a common hotspot mutant with dominant-negative and oncogenic activity. Using a novel iterative molecular docking-guided post-SELEX (systematic evolution of ligands by exponential enrichment) approach, we rationally engineer a high-performance DNA aptamer with improved affinity and specificity for p53-R175H. Leveraging this resulting aptamer as a binder for PROTACs, we successfully developed a selective p53-R175H degrader, named dp53m. dp53m induces the ubiquitin-proteasome-dependent degradation of p53-R175H while sparing wildtype p53. Importantly, dp53m demonstrates significant antitumor efficacy in p53-R175H-driven cancer cells both in vitro and in vivo, without toxicity. Moreover, dp53m significantly and synergistically improves the sensitivity of these cells to cisplatin, a commonly used chemotherapy drug. These findings provide evidence of the potential therapeutic value of dp53m in p53-R175H-driven cancers.
Collapse
Affiliation(s)
- Lingping Kong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fanlu Meng
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ping Zhou
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Ruixin Ge
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Xiaoshan Geng
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Zhihao Yang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Guo Li
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Linlin Zhang
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jing Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jinfeng Ma
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Cheng Dong
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jun Zhou
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China; Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Sijin Wu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215028, China.
| | - Diansheng Zhong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Songbo Xie
- Department of Ophthalmology, Tianjin Medical University General Hospital, Laboratory of Molecular Ophthalmology, Tianjin Key Laboratory of Ocular Trauma, Tianjin Medical University, Tianjin 300052, China.
| |
Collapse
|
10
|
Parviz M, Shokorlou YM, Heidarzadeh H. Structure of plasmonic multi spectral Apta sensor and analyzing of bulk and surface sensitivity. Sci Rep 2024; 14:13245. [PMID: 38853163 PMCID: PMC11163006 DOI: 10.1038/s41598-024-64249-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
In this work, a multispectral aptasensor structure, including a sub-layer and two side walls, was presented. The cells are positioned at the down and top of the structure, with the down cells oriented perpendicular to the walls and the top cells aligned parallel to the walls. The validity of the findings was verified by the utilization of a numerical simulation technique known as 3D Finite Difference Time Domain (FDTD). The biosensor under consideration exhibits sensitivities of 1093.7 nm/RIU, 754 nm/RIU, and 707.43 nm/RIU in mode III, mode II, and mode I, respectively. In the majority of instances, the quantity of analyte available is insufficient to coat the surface of the sensor thoroughly. Consequently, in this study, the evaluation of surface sensitivity was undertaken alongside bulk sensitivity. The surface sensitivity of the suggested structure for mode II in the sensor layer, with thicknesses of 10, 20, 30, and 70 nm, is measured to be 25, 78, 344, and 717.636 nm/RIU, respectively. Our design incorporates a unique arrangement of sub-layer and side walls, with cells positioned to maximize interaction with the target analyte. This innovative configuration, combined with Ag for its superior plasmonic properties, enables the detection of E. coli O157 with remarkable sensitivity.
Collapse
Affiliation(s)
- Mahya Parviz
- Department of Electrical Engineering, University of Kurdistan, Sanandaj, Kurdistan, Iran
| | - Younes Majd Shokorlou
- Department of Electrical and Computer Engineering, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hamid Heidarzadeh
- Department of Electrical and Computer Engineering, University of Mohaghegh Ardabili, Ardabil, Iran.
| |
Collapse
|
11
|
Ranjbari F, Nosrat A, Fathi F, Mohammadzadeh A. Surface plasmon resonance biosensors for early troponin detection. Clin Chim Acta 2024; 558:118670. [PMID: 38582245 DOI: 10.1016/j.cca.2024.118670] [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: 02/25/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Acute myocardial infarction (AMI) is one of the life-threatening causes that decrease blood flow to the heart, leading to increased mortality and related complications. Recently, the measure of blood concentration of cardiac biomarkers has been suggested to overcome the limitations of electrocardiography (ECG) analyses for early diagnosis of this disease. Troponins, especially cardiac troponin I and cardiac troponin T, with high sensitivity and specificity, are considered the gold standards in myocardial diagnosis. Recently, the use of new biosensors such as surface plasmon resonance (SPR) for early detection of these biomarkers has been greatly appreciated. Due to the rapid, sensitive, real-time, and label-free detection of SPR-based biosensors, they can be applied for selective and nonspecific absorption that is intended to be used as an in situ cardiac biosensor. Here, we exclusively discussed the updated developments of these valuable predictors for the possible occurrence of AMI detected by SPR.
Collapse
Affiliation(s)
- Faride Ranjbari
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Nosrat
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farzaneh Fathi
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alireza Mohammadzadeh
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
12
|
Li Y, Qian M, Liu Y, Qiu X. APPROACH: Sensitive Detection of Exosomal Biomarkers by Aptamer-Mediated Proximity Ligation Assay and Time-Resolved Förster Resonance Energy Transfer. BIOSENSORS 2024; 14:233. [PMID: 38785707 PMCID: PMC11117858 DOI: 10.3390/bios14050233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Exosomal biomarker detection holds great importance in the field of in vitro diagnostics, offering a non-invasive and highly sensitive approach for early disease detection and personalized treatment. Here, we proposed an "APPROACH" strategy, combining aptamer-mediated proximity ligation assay (PLA) with rolling circle amplification (RCA) and time-resolved Förster resonance energy transfer (TR-FRET) for the sensitive and semi-homogenous detection of exosomal biomarkers. PLA probes consisted of a cholesterol-conjugated oligonucleotide, which anchored to the membrane of an exosome, and a specific aptamer oligonucleotide that recognized a target protein of the exosome; the proximal binding of pairs of PLA probes to the same exosome positioned the oligonucleotides in the vicinity of each other, guiding the hybridization and ligation of two subsequently added backbone and connector oligonucleotides to form a circular DNA molecule. Circular DNA formed from PLA underwent rolling circle amplification (RCA) for signal amplification, and the resulting RCA products were subsequently quantified by TR-FRET. The limits of detection provided by APPROACH for the exosomal biomarkers CD63, PD-L1, and HER2 were 0.46 ng∙μL-1, 0.77 ng∙μL-1, and 1.1 ng∙μL-1, respectively, demonstrating excellent analytical performance with high sensitivity and quantification accuracy. Furthermore, the strategy afforded sensitive detection of exosomal CD63 with a LOD of 1.56 ng∙μL-1 in complex biological matrices, which underscored its anti-interference capability and potential for in vitro detection. The proposed strategy demonstrates wide-ranging applicability in quantifying diverse exosomal biomarkers while exhibiting robust analytical characteristics, including high sensitivity and accuracy.
Collapse
Affiliation(s)
- Ying Li
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.L.); (M.Q.)
| | - Meiqi Qian
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.L.); (M.Q.)
| | | | - Xue Qiu
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.L.); (M.Q.)
| |
Collapse
|
13
|
Araújo D, Silva AR, Fernandes R, Serra P, Barros MM, Campos AM, Oliveira R, Silva S, Almeida C, Castro J. Emerging Approaches for Mitigating Biofilm-Formation-Associated Infections in Farm, Wild, and Companion Animals. Pathogens 2024; 13:320. [PMID: 38668275 PMCID: PMC11054384 DOI: 10.3390/pathogens13040320] [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: 03/15/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
The importance of addressing the problem of biofilms in farm, wild, and companion animals lies in their pervasive impact on animal health and welfare. Biofilms, as resilient communities of microorganisms, pose a persistent challenge in causing infections and complicating treatment strategies. Recognizing and understanding the importance of mitigating biofilm formation is critical to ensuring the welfare of animals in a variety of settings, from farms to the wild and companion animals. Effectively addressing this issue not only improves the overall health of individual animals, but also contributes to the broader goals of sustainable agriculture, wildlife conservation, and responsible pet ownership. This review examines the current understanding of biofilm formation in animal diseases and elucidates the complex processes involved. Recognizing the limitations of traditional antibiotic treatments, mechanisms of resistance associated with biofilms are explored. The focus is on alternative therapeutic strategies to control biofilm, with illuminating case studies providing valuable context and practical insights. In conclusion, the review highlights the importance of exploring emerging approaches to mitigate biofilm formation in animals. It consolidates existing knowledge, highlights gaps in understanding, and encourages further research to address this critical facet of animal health. The comprehensive perspective provided by this review serves as a foundation for future investigations and interventions to improve the management of biofilm-associated infections in diverse animal populations.
Collapse
Affiliation(s)
- Daniela Araújo
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
| | - Ana Rita Silva
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Rúben Fernandes
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Patrícia Serra
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Maria Margarida Barros
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CECAV—Veterinary and Animal Research Centre, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Ana Maria Campos
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
| | - Ricardo Oliveira
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sónia Silva
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
| | - Carina Almeida
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- AliCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana Castro
- INIAV—National Institute for Agrarian and Veterinarian Research, Rua dos Lagidos, 4485-655 Vila do Conde, Portugal; (A.R.S.); (R.F.); (P.S.); (M.M.B.); (A.M.C.); (R.O.); (S.S.); (C.A.)
- CEB—Centre of Biological Engineering Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
14
|
Cai T, Chen M, Yang J, Tang C, Lu X, Wei Z, Jiang H, Hou Y, Zhao J, Yu P. An AuNPs-based electrochemical aptasensor for the detection of 25-hydroxy vitamin D 3. ANAL SCI 2024; 40:599-607. [PMID: 38190076 DOI: 10.1007/s44211-023-00489-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024]
Abstract
Vitamin D3 (VD3) is the main form of vitamin D and an essential nutrient for maintaining human life. Currently, traditional methods for detecting 25-hydroxyvitamin D3(25(OH)D3) are complex and expensive. In this study, we constructed an accurate, sensitive, simple, and cost-effective label-free biosensor based on an aptamer for the detection of 25(OH)D3. The aptamer-modified sulfhydryl adopted self-assembly as a way to stably immobilize at the glassy carbon electrode (GCE) surface modified by gold nanoparticles (AuNPs). Upon 25(OH)D3 binding to the aptamer, the complexes inhibit electron transfer at the electrode surface, leading to reduced [Fe(CN)6]3-/4- redox peak current. Consequently, the quantity of 25(OH)D3 that interacts with the electrode-bound aptamer correlates with the observed electric current response values. The Aptamer/AuNPs/GCE aptasensor achieved direct and highly sensitive detection of 25(OH)D3 over a wide concentration range (1.0-1000 nM), with a limit of detection of 1.0 nM. At the same time, other molecules with a similar structure, such as 25(OH)D2, Vitamin D3, and Vitamin D2, had lower response interference than 25(OH)D3. Therefore, this biosensor has great potential to become a portable diagnostic device for 25(OH)D3.
Collapse
Affiliation(s)
- Tongji Cai
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Meilun Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jie Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Chunhua Tang
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Xiaoling Lu
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Zheng Wei
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Hanbing Jiang
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yucui Hou
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jia Zhao
- Changsha Cinotohi Technology Co., Ltd, No. 601, North Dongfanghong Road, Changsha, 410013, Hunan, China
| | - Peng Yu
- Xiangya School of Pharmaceutical Sciences, Central South University, No. 172, Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
15
|
Dinesen A, Andersen VL, Elkhashab M, Pilati D, Bech P, Fuchs E, Samuelsen TR, Winther A, Cai Y, Märcher A, Wall A, Omer M, Nielsen JS, Chudasama V, Baker JR, Gothelf KV, Wengel J, Kjems J, Howard KA. An Albumin-Holliday Junction Biomolecular Modular Design for Programmable Multifunctionality and Prolonged Circulation. Bioconjug Chem 2024; 35:214-222. [PMID: 38231391 PMCID: PMC10886128 DOI: 10.1021/acs.bioconjchem.3c00491] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/18/2024]
Abstract
Combinatorial properties such as long-circulation and site- and cell-specific engagement need to be built into the design of advanced drug delivery systems to maximize drug payload efficacy. This work introduces a four-stranded oligonucleotide Holliday Junction (HJ) motif bearing functional moieties covalently conjugated to recombinant human albumin (rHA) to give a "plug-and-play" rHA-HJ multifunctional biomolecular assembly with extended circulation. Electrophoretic gel-shift assays show successful functionalization and purity of the individual high-performance liquid chromatography-purified modules as well as efficient assembly of the rHA-HJ construct. Inclusion of an epidermal growth factor receptor (EGFR)-targeting nanobody module facilitates specific binding to EGFR-expressing cells resulting in approximately 150-fold increased fluorescence intensity determined by flow cytometric analysis compared to assemblies absent of nanobody inclusion. A cellular recycling assay demonstrated retained albumin-neonatal Fc receptor (FcRn) binding affinity and accompanying FcRn-driven cellular recycling. This translated to a 4-fold circulatory half-life extension (2.2 and 0.55 h, for the rHA-HJ and HJ, respectively) in a double transgenic humanized FcRn/albumin mouse. This work introduces a novel biomolecular albumin-nucleic acid construct with extended circulatory half-life and programmable multifunctionality due to its modular design.
Collapse
Affiliation(s)
- Anders Dinesen
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Veronica L. Andersen
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Marwa Elkhashab
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Diego Pilati
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Pernille Bech
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Elisabeth Fuchs
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Torbjørn R. Samuelsen
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Alexander Winther
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Yunpeng Cai
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Anders Märcher
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Archie Wall
- Department
of Chemistry, University College London, London WC1H 0AJ, U.K.
| | - Marjan Omer
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Jesper S. Nielsen
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Vijay Chudasama
- Department
of Chemistry, University College London, London WC1H 0AJ, U.K.
| | - James R. Baker
- Department
of Chemistry, University College London, London WC1H 0AJ, U.K.
| | - Kurt V. Gothelf
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jesper Wengel
- Nucleic
Acid Center, Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Jørgen Kjems
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| | - Kenneth A. Howard
- Interdisciplinary
Nanoscience Center (iNANO) and Department of Molecular Biology and
Genetics, Aarhus University, DK-8000 Aarhus
C, Denmark
| |
Collapse
|
16
|
Wang C, Wang T, Gao Y, Tao Q, Ye W, Jia Y, Zhao X, Zhang B, Zhang Z. Multiplexed immunosensing of cancer biomarkers on a split-float-gate graphene transistor microfluidic biochip. LAB ON A CHIP 2024; 24:317-326. [PMID: 38087953 DOI: 10.1039/d3lc00709j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
This work reports the development of a novel microfluidic biosensor using a graphene field-effect transistor (GFET) design for the parallel label-free analysis of multiple biomarkers. Overcoming the persistent challenge of constructing μm2-sized FET sensitive interfaces that incorporate multiple receptors, we implement a split-float-gate structure that enables the manipulation of multiplexed biochemical functionalization using microfluidic channels. Immunoaffinity biosensing experiments are conducted using the mixture samples containing three liver cancer biomarkers, carcinoembryonic antigen (CEA), α-fetoprotein (AFP), and parathyroid hormone (PTH). The results demonstrate the capability of our label-free biochip to quantitatively detect multiple target biomarkers simultaneously by observing the kinetics in 10 minutes, with the detection limit levels in the nanomolar range. This microfluidic biosensor provides a valuable analytical tool for rapid multi-target biosensing, which can be potentially utilized for domiciliary tests of cancer screening and prognosis, obviating the need for sophisticated instruments and professional operations in hospitals.
Collapse
Affiliation(s)
- Cheng Wang
- Tianjin Key Laboratory of Wireless Mobile Communications and Power Transmission, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China.
- Department of Intelligence Science and Technology, College of Artificial Intelligence, Tianjin Normal University, Tianjin 300387, China
| | - Tao Wang
- Tianjin Key Laboratory of Wireless Mobile Communications and Power Transmission, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China.
- Department of Communication Engineering, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China
| | - Yujing Gao
- Tianjin Key Laboratory of Wireless Mobile Communications and Power Transmission, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China.
- Department of Intelligence Science and Technology, College of Artificial Intelligence, Tianjin Normal University, Tianjin 300387, China
| | - Qiya Tao
- Tianjin Key Laboratory of Wireless Mobile Communications and Power Transmission, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China.
- Department of Communication Engineering, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China
| | - Weixiang Ye
- Center for Theoretical Physics, Hainan University, Haikou 570228, China.
- Department of Physics, School of Physical Science and Optoelectrical Engineering, Hainan University, Haikou 570228, China
| | - Yuan Jia
- Industrialization Center of Micro/Nano ICs and Devices, Sino-German College of Intelligent Manufacturing, Shenzhen Technology University, Shenzhen 518118, China.
| | - Xiaonan Zhao
- Tianjin Key Laboratory of Wireless Mobile Communications and Power Transmission, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China.
- Department of Communication Engineering, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China
| | - Bo Zhang
- Tianjin Key Laboratory of Wireless Mobile Communications and Power Transmission, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China.
- Department of Communication Engineering, College of Electronic and Communication Engineering, Tianjin Normal University, Tianjin 300387, China
| | - Zhixing Zhang
- Industrialization Center of Micro/Nano ICs and Devices, Sino-German College of Intelligent Manufacturing, Shenzhen Technology University, Shenzhen 518118, China.
| |
Collapse
|
17
|
Guo ZY, Tang Y, Cheng YC. Exosomes as Targeted Delivery Drug System: Advances in Exosome Loading, Surface Functionalization and Potential for Clinical Application. Curr Drug Deliv 2024; 21:473-487. [PMID: 35702803 DOI: 10.2174/1567201819666220613150814] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022]
Abstract
Exosomes are subtypes of vesicles secreted by almost all cells and can play an important role in intercellular communication. They contain various proteins, lipids, nucleic acids and other natural substances from their metrocytes. Exosomes are expected to be a new generation of drug delivery systems due to their low immunogenicity, high potential to transfer bioactive substances and biocompatibility. However, exosomes themselves are not highly targeted, it is necessary to develop new surface modification techniques and targeted drug delivery strategies, which are the focus of drug delivery research. In this review, we introduced the biogenesis of exosomes and their role in intercellular communication. We listed various advanced exosome drug-loading techniques. Emphatically, we summarized different exosome surface modification techniques and targeted drug delivery strategies. In addition, we discussed the application of exosomes in vaccines and briefly introduced milk exosomes. Finally, we clarified the clinical application prospects and shortcomings of exosomes.
Collapse
Affiliation(s)
- Zun Y Guo
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing 211198, P.R. China
| | - Yue Tang
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing 211198, P.R. China
| | - Yi C Cheng
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing 211198, P.R. China
| |
Collapse
|
18
|
Ma T, Huang K, Cheng N. Recent Advances in Nanozyme-Mediated Strategies for Pathogen Detection and Control. Int J Mol Sci 2023; 24:13342. [PMID: 37686145 PMCID: PMC10487713 DOI: 10.3390/ijms241713342] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/13/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Pathogen detection and control have long presented formidable challenges in the domains of medicine and public health. This review paper underscores the potential of nanozymes as emerging bio-mimetic enzymes that hold promise in effectively tackling these challenges. The key features and advantages of nanozymes are introduced, encompassing their comparable catalytic activity to natural enzymes, enhanced stability and reliability, cost effectiveness, and straightforward preparation methods. Subsequently, the paper delves into the detailed utilization of nanozymes for pathogen detection. This includes their application as biosensors, facilitating rapid and sensitive identification of diverse pathogens, including bacteria, viruses, and plasmodium. Furthermore, the paper explores strategies employing nanozymes for pathogen control, such as the regulation of reactive oxygen species (ROS), HOBr/Cl regulation, and clearance of extracellular DNA to impede pathogen growth and transmission. The review underscores the vast potential of nanozymes in pathogen detection and control through numerous specific examples and case studies. The authors highlight the efficiency, rapidity, and specificity of pathogen detection achieved with nanozymes, employing various strategies. They also demonstrate the feasibility of nanozymes in hindering pathogen growth and transmission. These innovative approaches employing nanozymes are projected to provide novel options for early disease diagnoses, treatment, and prevention. Through a comprehensive discourse on the characteristics and advantages of nanozymes, as well as diverse application approaches, this paper serves as a crucial reference and guide for further research and development in nanozyme technology. The expectation is that such advancements will significantly contribute to enhancing disease control measures and improving public health outcomes.
Collapse
Affiliation(s)
- Tianyi Ma
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.M.); (K.H.)
| | - Kunlun Huang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.M.); (K.H.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Nan Cheng
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.M.); (K.H.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| |
Collapse
|
19
|
Bertrand P. Aptamers Targeting the PD-1/PD-L1 Axis: A Perspective. J Med Chem 2023; 66:10878-10888. [PMID: 37561598 DOI: 10.1021/acs.jmedchem.3c00551] [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: 08/12/2023]
Abstract
Aptamers have emerged in recent years as alternatives to antibodies or small molecules to interfere with the immune check points by blocking the PD-1/PD-L1 interactions and represent an interesting perspective for immuno-oncology. Aptamers are RNA or DNA nucleotides able to bind to a target with high affinity, with the target ranging from small molecules to proteins and up to cells. Aptamers are identified by the SELEX method that can be modified for specific purposes. The range of applications of aptamers covers therapy as well as new alternative assay technologies similar to ELISA. Aptamers' limited plasma stability can be managed using delivery strategies. The goal of this Perspective is to give an overview of the current development of aptamers targeting the most studied immune checkpoint modulators, PD-1 and PD-L1, and analogous strategies with aptamers for other immuno-related targets.
Collapse
Affiliation(s)
- Philippe Bertrand
- University of Poitiers, IC2MP UMR 7285 CNRS, 4 rue Michel Brunet B27, TSA 51106, 86073 Poitiers cedex 9, France
| |
Collapse
|
20
|
Ling M, Cardle II, Song K, Yan AJ, Kacherovsky N, Jensen MC, Pun SH. Aptamer-Based Chromatographic Methods for Efficient and Economical Separation of Leukocyte Populations. ACS Biomater Sci Eng 2023; 9:5062-5071. [PMID: 37467493 PMCID: PMC11016351 DOI: 10.1021/acsbiomaterials.3c00651] [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] [Indexed: 07/21/2023]
Abstract
The manufacturing process of chimeric antigen receptor T cell therapies includes isolation systems that provide pure T cells. Current magnetic-activated cell sorting and immunoaffinity chromatography methods produce desired cells with high purity and yield but require expensive equipment and reagents and involve time-consuming incubation steps. Here, we demonstrate that aptamers can be employed in a continuous-flow resin platform for both depletion of monocytes and selection of CD8+ T cells from peripheral blood mononuclear cells at low cost with high purity and throughput. Aptamer-mediated cell selection could potentially enable fully synthetic, traceless isolations of leukocyte subsets from a single isolation system.
Collapse
Affiliation(s)
- Melissa Ling
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195
| | - Ian I. Cardle
- Department of Bioengineering, University of Washington, Seattle, WA 98195
- Seattle Children’s Therapeutics, Seattle, WA 98101
| | - Kefan Song
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Alexander J. Yan
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Nataly Kacherovsky
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | | | - Suzie H. Pun
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98195
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| |
Collapse
|
21
|
Xiong H, Li P, Cun F, Chen H, Kong J. Methylene-Blue-Encapsulated Metal-Organic-Framework-Based Electrochemical POCT Platform for Multiple Detection of Heavy Metal Ions in Milk. BIOSENSORS 2023; 13:783. [PMID: 37622869 PMCID: PMC10452309 DOI: 10.3390/bios13080783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023]
Abstract
Considering the high risk of heavy metal ions (HMIs) transferring through the food chain and accumulating in milk, a flexible and facile point-of-care testing (POCT) platform is urgently needed for the accurate, sensitive, and highly selective on-site quantification of multiple HMIs in milk. In this work, a cost-effective disk with six screen-printed electrodes (SPEs) was designed for hand-held electrochemical detection. Metal organic frameworks (MOFs) were adopted to amplify and enhance the electrochemical signals of methylene blue (MB). Using differential pulse voltammetry (DPV) methods, low limits of detection for four HMIs (Cd2+, 0.039 ppb; Hg2+, 0.039 ppb; Pb2+, 0.073 ppb; and As3+, 0.022 ppb) were achieved within four minutes. Moreover, the quantitative POCT system was applied to milk samples. The advantages of low cost, ease of on-site implementation, fast response, and accuracy allow for the POCT platform to be used in practical monitoring applications for the quantitation of multiple HMIs in milk samples.
Collapse
Affiliation(s)
| | | | | | - Hui Chen
- Department of Chemistry, Fudan University, Shanghai 200438, China
| | - Jilie Kong
- Department of Chemistry, Fudan University, Shanghai 200438, China
| |
Collapse
|
22
|
Liu X, Li F, Dong Z, Gu C, Mao D, Chen J, Luo L, Huang Y, Xiao J, Li Z, Liu Z, Yang Y. Metal-polyDNA nanoparticles reconstruct osteoporotic microenvironment for enhanced osteoporosis treatment. SCIENCE ADVANCES 2023; 9:eadf3329. [PMID: 37531423 PMCID: PMC10396296 DOI: 10.1126/sciadv.adf3329] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
Current clinical approaches to osteoporosis primarily target osteoclast biology, overlooking the synergistic role of bone cells, immune cells, cytokines, and inorganic components in creating an abnormal osteoporotic microenvironment. Here, metal-polyDNA nanoparticles (Ca-polyCpG MDNs) composed of Ca2+ and ultralong single-stranded CpG sequences were developed to reconstruct the osteoporotic microenvironment and suppress osteoporosis. Ca-polyCpG MDNs can neutralize osteoclast-secreted hydrogen ions, provide calcium repletion, promote remineralization, and repair bone defects. Besides, the immune-adjuvant polyCpG in MDNs could induce the secretion of osteoclastogenesis inhibitor interleukin-12 and reduce the expression of osteoclast function effector protein to inhibit osteoclast differentiation, further reducing osteoclast-mediated bone resorption. PPi4- generated during the rolling circle amplification reaction acts as bisphosphonate analog and enhances bone targeting of Ca-polyCpG MDNs. In ovariectomized mouse and rabbit models, Ca-polyCpG MDNs prevented bone resorption and promoted bone repair by restoring the osteoporotic microenvironment, providing valuable insights into osteoporosis therapy.
Collapse
Affiliation(s)
- Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fan Li
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Ziliang Dong
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chao Gu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dongsheng Mao
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jingqi Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Luo
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuting Huang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Xiao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Zhanchun Li
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhuang Liu
- Department of Orthopaedic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
23
|
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
|
24
|
Srinivasan S, Ranganathan V, McConnell EM, Murari BM, DeRosa MC. Aptamer-based colorimetric and lateral flow assay approaches for the detection of toxic metal ions, thallium(i) and lead(ii). RSC Adv 2023; 13:20040-20049. [PMID: 37409036 PMCID: PMC10318611 DOI: 10.1039/d3ra01658g] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/14/2023] [Indexed: 07/07/2023] Open
Abstract
Thallium(i) and lead(ii) ions are heavy metals and extremely toxic. These metals are environmental pollutants, posing a severe risk to the environment and human health. In this study, two approaches were examined using aptamer and nanomaterial-based conjugates for thallium and lead detection. The first approach utilized an in-solution adsorption-desorption approach to develop colorimetric aptasensors for the detection of thallium(i) and lead(ii) using gold or silver nanoparticles. The second approach was the development of lateral flow assays, and their performance was tested with thallium (limit of detection is 7.4 μM) and lead ion (limit of detection is 6.6 nM) spiked into real samples. The approaches assessed are rapid, inexpensive, and time efficient with the potential to become the basis for future biosensor devices.
Collapse
Affiliation(s)
- Sathya Srinivasan
- Department of Chemistry, Carleton University 1125 Colonel By Drive Ottawa ON K1S 5B6 Canada +1-613-520-2600 ext. 4388
- Department of Biotechnology, School of Bioscience and Technology VIT Vellore 632 104 TN India
| | - Velu Ranganathan
- Department of Chemistry, Carleton University 1125 Colonel By Drive Ottawa ON K1S 5B6 Canada +1-613-520-2600 ext. 4388
| | - Erin M McConnell
- Department of Chemistry, Carleton University 1125 Colonel By Drive Ottawa ON K1S 5B6 Canada +1-613-520-2600 ext. 4388
| | - Bhaskar Mohan Murari
- Department of Sensor and Biomedical Technology, School of Electronics Engineering VIT Vellore 632 104 TN India
| | - Maria C DeRosa
- Department of Chemistry, Carleton University 1125 Colonel By Drive Ottawa ON K1S 5B6 Canada +1-613-520-2600 ext. 4388
| |
Collapse
|
25
|
Wang J, Chen D, Huang W, Yang N, Yuan Q, Yang Y. Aptamer-functionalized field-effect transistor biosensors for disease diagnosis and environmental monitoring. EXPLORATION (BEIJING, CHINA) 2023; 3:20210027. [PMID: 37933385 PMCID: PMC10624392 DOI: 10.1002/exp.20210027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/10/2023] [Indexed: 11/08/2023]
Abstract
Nano-biosensors that are composed of recognition molecules and nanomaterials have been extensively utilized in disease diagnosis, health management, and environmental monitoring. As a type of nano-biosensors, molecular specificity field-effect transistor (FET) biosensors with signal amplification capability exhibit prominent advantages including fast response speed, ease of miniaturization, and integration, promising their high sensitivity for molecules detection and identification. With intrinsic characteristics of high stability and structural tunability, aptamer has become one of the most commonly applied biological recognition units in the FET sensing fields. This review summarizes the recent progress of FET biosensors based on aptamer functionalized nanomaterials in medical diagnosis and environmental monitoring. The structure, sensing principles, preparation methods, and functionalization strategies of aptamer modified FET biosensors were comprehensively summarized. The relationship between structure and sensing performance of FET biosensors was reviewed. Furthermore, the challenges and future perspectives of FET biosensors were also discussed, so as to provide support for the future development of efficient healthcare management and environmental monitoring devices.
Collapse
Affiliation(s)
- Jingfeng Wang
- College of Chemistry and Molecular Sciences, Institute of Molecular MedicineRenmin Hospital of Wuhan University, School of Microelectronics, Wuhan UniversityWuhanChina
| | - Duo Chen
- College of Chemistry and Molecular Sciences, Institute of Molecular MedicineRenmin Hospital of Wuhan University, School of Microelectronics, Wuhan UniversityWuhanChina
| | - Wanting Huang
- College of Chemistry and Molecular Sciences, Institute of Molecular MedicineRenmin Hospital of Wuhan University, School of Microelectronics, Wuhan UniversityWuhanChina
| | - Nianjun Yang
- Department of Chemistry, Insititute of Materials ResearchHasselt UniversityHasseltBelgium
| | - Quan Yuan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical EngineeringHunan UniversityChangshaChina
| | - Yanbing Yang
- College of Chemistry and Molecular Sciences, Institute of Molecular MedicineRenmin Hospital of Wuhan University, School of Microelectronics, Wuhan UniversityWuhanChina
| |
Collapse
|
26
|
Gao J, Yang L, Lei S, Zhou F, Nie H, Peng B, Xu T, Chen X, Yang X, Sheng C, Rao Y, Pu K, Jin J, Xu Z, Yu H. Stimuli-activatable PROTACs for precise protein degradation and cancer therapy. Sci Bull (Beijing) 2023; 68:1069-1085. [PMID: 37169612 DOI: 10.1016/j.scib.2023.04.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/28/2023] [Accepted: 04/21/2023] [Indexed: 05/13/2023]
Abstract
The proteolysis targeting chimeras (PROTACs) approach has attracted extensive attention in the past decade, which represents an emerging therapeutic modality with the potential to tackle disease-causing proteins that are historically challengeable for conventional small molecular inhibitors. PROTAC harnesses the endogenic E3 ubiquitin ligase to degrade protein of interest (POI) via ubiquitin-proteasome system in a cycle-catalytic manner. The event-driven pharmacology of PROTAC is poised to pursue those targets that are conventionally undruggable, which enormously extends the space of drug development. Furthermore, PROTAC has the potential to address drug resistance of small molecular inhibitors by degrading the whole POI. Nevertheless, PROTACs display high-efficiency and always-on properties to degrade POI, they may cause severe side effects due to an "on-target but off-tissue" protein degradation profile at the undesirable tissues and cells. Given that, the stimuli-activatable PROTAC prodrugs have been recently exploited to confine precise protein degradation of the favorable targets, which may conquer the adverse effects of PROTAC due to uncontrollable protein degradation. Herein, we summarized the cutting-edge advances of the stimuli-activatable PROTAC prodrugs. We also overviewed the progress of PROTAC prodrug-based nanomedicine to improve PROTAC delivery to the tumors and precise POI degradation in the targeted cells.
Collapse
Affiliation(s)
- Jing Gao
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lei Yang
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shumin Lei
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Feng Zhou
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huijun Nie
- Center of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bo Peng
- Information Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tianfeng Xu
- Center of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaohua Chen
- Center of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Xiaobao Yang
- Gluetacs Therapeutics (Shanghai) Co., Ltd. Shanghai 201306, China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yu Rao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences, Oncological Sciences and Neuroscience, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York NY 10029, USA
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| | - Haijun Yu
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
27
|
Neff CP, Cikara M, Geiss BJ, Thomas Caltagirone G, Liao A, Atif SM, Macdonald B, Schaden R. Nucleocapsid protein binding DNA aptamers for detection of SARS-COV-2. CURRENT RESEARCH IN BIOTECHNOLOGY 2023; 5:100132. [PMID: 37275459 PMCID: PMC10223630 DOI: 10.1016/j.crbiot.2023.100132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/07/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus (SARS-CoV-2) has infected millions of individuals and continues to be a major health concern worldwide. While reverse transcription-polymerase chain reaction remains a reliable method for detecting infections, limitations of this technology, particularly cost and the requirement of a dedicated laboratory, prevent rapid viral monitoring. Antigen tests filled this need to some extent but with limitations including sensitivity and specificity, particularly against emerging variants of concern. Here, we developed aptamers against the SARS-CoV-2 Nucleocapsid protein to complement or replace antibodies in antigen detection assays. As detection reagents in ELISA-like assays, our DNA aptamers were able to detect as low as 150 pg/mL of the protein and under 150 k copies of inactivated SARS-CoV-2 Wuhan Alpha strain in viral transport medium with little cross-reactivity to other human coronaviruses (HCoVs). Further, our aptamers were reselected against the SARS-CoV-2 Omicron variant of concern, and we found two sequences that had a more than two-fold increase in signal compared to our original aptamers when used as detection reagents against protein from the Omicron strain. These findings illustrate the use of aptamers as promising alternative detection reagents that may translate for use in current tests and our findings validate the method for the reselection of aptamers against emerging viral strains.
Collapse
Affiliation(s)
- Charles P Neff
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mile Cikara
- Precision Medicine Architects, LLC, PO Box 148, Wellington, CO 80549, United States
| | - Brian J Geiss
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Albert Liao
- Aptagen, LLC, 250 North Main Street, Jacobus, PA 17407, USA
| | - Shaikh M Atif
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bradley Macdonald
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Richard Schaden
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
28
|
Yang LF, Ling M, Kacherovsky N, Pun SH. Aptamers 101: aptamer discovery and in vitro applications in biosensors and separations. Chem Sci 2023; 14:4961-4978. [PMID: 37206388 PMCID: PMC10189874 DOI: 10.1039/d3sc00439b] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/14/2023] [Indexed: 05/21/2023] Open
Abstract
Aptamers are single-stranded nucleic acids that bind and recognize targets much like antibodies. Recently, aptamers have garnered increased interest due to their unique properties, including inexpensive production, simple chemical modification, and long-term stability. At the same time, aptamers possess similar binding affinity and specificity as their protein counterpart. In this review, we discuss the aptamer discovery process as well as aptamer applications to biosensors and separations. In the discovery section, we describe the major steps of the library selection process for aptamers, called systematic evolution of ligands by exponential enrichment (SELEX). We highlight common approaches and emerging strategies in SELEX, from starting library selection to aptamer-target binding characterization. In the applications section, we first evaluate recently developed aptamer biosensors for SARS-CoV-2 virus detection, including electrochemical aptamer-based sensors and lateral flow assays. Then we discuss aptamer-based separations for partitioning different molecules or cell types, especially for purifying T cell subsets for therapeutic applications. Overall, aptamers are promising biomolecular tools and the aptamer field is primed for expansion in biosensing and cell separation.
Collapse
Affiliation(s)
- Lucy F Yang
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington Seattle Washington USA
| | - Melissa Ling
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington Seattle Washington USA
| | - Nataly Kacherovsky
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington Seattle Washington USA
| | - Suzie H Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington Seattle Washington USA
| |
Collapse
|
29
|
Goldstein I, Alyas S, Asghar W, Ilyas A. Biosensors for the Isolation and Detection of Circulating Tumor Cells (CTCs) in Point-of-Care Settings. MICROMACHINES 2023; 14:mi14051035. [PMID: 37241658 DOI: 10.3390/mi14051035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023]
Abstract
Circulating tumor cells (CTCs) are cells that have been shed from tumors and circulate in the bloodstream. These cells can also be responsible for further metastases and the spread of cancer. Taking a closer look and analyzing CTCs through what has come to be known as "liquid biopsy" has immense potential to further researchers' understanding of cancer biology. However, CTCs are very sparse and are therefore difficult to detect and capture. To combat this issue, researchers have attempted to create devices, assays, and further techniques to successfully isolate CTCs for analysis. In this work, new and existing biosensing techniques for CTC isolation, detection, and release/detachment are discussed and compared to evaluate their efficacy, specificity, and cost. Here, we specifically aim to evaluate and identify the potential success of these techniques and devices in point-of-care (POC) settings.
Collapse
Affiliation(s)
- Isaac Goldstein
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- Paul D. Schreiber High School, Port Washington, NY 11050, USA
| | - Sobia Alyas
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore 54000, Pakistan
| | - Waseem Asghar
- Department of Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Azhar Ilyas
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA
- Department of Electrical and Computer Engineering, New York Institute of Technology, Old Westbury, NY 11568, USA
| |
Collapse
|
30
|
Sincere NI, Anand K, Ashique S, Yang J, You C. PROTACs: Emerging Targeted Protein Degradation Approaches for Advanced Druggable Strategies. Molecules 2023; 28:molecules28104014. [PMID: 37241755 DOI: 10.3390/molecules28104014] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/22/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
A potential therapeutic strategy to treat conditions brought on by the aberrant production of a disease-causing protein is emerging for targeted protein breakdown using the PROTACs technology. Few medications now in use are tiny, component-based and utilize occupancy-driven pharmacology (MOA), which inhibits protein function for a short period of time to temporarily alter it. By utilizing an event-driven MOA, the proteolysis-targeting chimeras (PROTACs) technology introduces a revolutionary tactic. Small-molecule-based heterobifunctional PROTACs hijack the ubiquitin-proteasome system to trigger the degradation of the target protein. The main challenge PROTAC's development facing now is to find potent, tissue- and cell-specific PROTAC compounds with favorable drug-likeness and standard safety measures. The ways to increase the efficacy and selectivity of PROTACs are the main focus of this review. In this review, we have highlighted the most important discoveries related to the degradation of proteins by PROTACs, new targeted approaches to boost proteolysis' effectiveness and development, and promising future directions in medicine.
Collapse
Affiliation(s)
- Nuwayo Ishimwe Sincere
- Laboratory Medicine Center, Lanzhou University Second Hospital, The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Sumel Ashique
- Department of Pharmaceutics, Bharat Institute of Technology (BIT), School of Pharmacy, Meerut 250103, India
| | - Jing Yang
- Laboratory Medicine Center, Lanzhou University Second Hospital, The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Chongge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
31
|
Pan Z, Zhu H, Zhang Y, Liao Q, Sun Y, Wu E, Wang Y, Shi K, Zhang Y, Chen L, Ye M, Wu W. Development of Uveal Melanoma-Specific Aptamer for Potential Biomarker Discovery and Targeted Drug Delivery. Anal Chem 2023; 95:5095-5108. [PMID: 36812415 DOI: 10.1021/acs.analchem.3c00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. However, challenges in early diagnosis, high risk of liver metastasis, and lack of effective targeted therapy lead to poor prognosis and high mortality of UM. Therefore, generating an effective molecular tool for UM diagnosis and targeted treatment is of great significance. In this study, a UM-specific DNA aptamer, PZ-1, was successfully developed, which could specifically distinguish molecular differences between UM cells and noncancerous cells with nanomolar-range affinity and presented excellent recognition ability for UM in vivo and clinical UM tissues. Subsequently, the binding target of PZ-1 on UM cells was identified as JUP (junction plakoglobin) protein, which held great potential as a biomarker and therapeutic target for UM. Meanwhile, the strong stability and internalization capacity of PZ-1 were also determined, and a UM-specific aptamer-guided "nanoship" was engineered to load and selectively release doxorubicin (Dox) to targeted UM cells, with lower toxicity to nontumor cells. Taken together, the UM-specific aptamer PZ-1 could serve as a molecular tool to discover the potential biomarker for UM and to achieve the targeted therapy of UM.
Collapse
Affiliation(s)
- Zhaoqi Pan
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Hui Zhu
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Yibin Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410082, China
| | - Qianling Liao
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yiping Sun
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ende Wu
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Yanan Wang
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Kesi Shi
- The second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Yikui Zhang
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Lu Chen
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Wencan Wu
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| |
Collapse
|
32
|
Suliman Maashi M. CRISPR/Cas-based Aptasensor as an Innovative Sensing Approaches for Food Safety Analysis: Recent Progresses and New Horizons. Crit Rev Anal Chem 2023; 54:2599-2617. [PMID: 36940173 DOI: 10.1080/10408347.2023.2188955] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
Food safety is one of the greatest public problems occurring around the world. Chemical, physical, and microbiological hazards could lead to food safety problems, which might occur at all stages of the supply chain. To tackle food safety problems and protect consumer health, specific, accurate, and rapid diagnosis techniques meeting various requirements are the imperative measures to ensure food safety. CRISPR-Cas system, a novel emerging technology, is effectively repurposed in (bio)sensing and has shown a tremendous capability to develop on-site and portable diagnostic methods with high specificity and sensitivity. Among numerous existing CRISPR/Cas systems, CRISPR/Cas13a and CRISPR/Cas12a are extensively employed in the design of biosensors, owing to their ability to cleave both non-target and target sequences. However, the specificity limitation in CRISPR/Cas has hindered its progress. Nowadays, nucleic acid aptamers recognized for their specificity and high-affinity characteristics for their analytes are incorporated into CRISPR/Cas systems. With the benefits of reproducibility, high durability, portability, facile operation, and cost-effectiveness, CRISPR/Cas-based aptasensing approaches are an ideal choice for fabricating highly specific point-of-need analytical tools with enhanced response signals. In the current study, we explore some of the most recent progress in the CRISPR/Cas-mediated aptasensors for detecting food risk factors including veterinary drugs, pesticide residues, pathogens, mycotoxins, heavy metals, illegal additives, food additives, and other contaminants. The nanomaterial engineering support with CRISPR/Cas aptasensors is also signified to achieve a hopeful perspective to provide new straightforward test kits toward trace amounts of different contaminants encountered in food samples.
Collapse
Affiliation(s)
- Marwah Suliman Maashi
- Medical Laboratory Science Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah, Saudi Arabia
| |
Collapse
|
33
|
Chu X, Du X, Yang L, Wang Z, Zhang Y, Wang X, Dai L, Zhang J, Liu J, Zhang N, Zhao Y, Gu H. Targeting Tumor Necrosis Factor Receptor 1 with Selected Aptamers for Anti-Inflammatory Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:11599-11608. [PMID: 36812453 DOI: 10.1021/acsami.3c00131] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Tumor necrosis factor-α (TNFα) inhibitors are widely used in treating autoimmune diseases like rheumatoid arthritis (RA). These inhibitors can presumably alleviate RA symptoms by blocking TNFα-TNF receptor 1 (TNFR1)-mediated pro-inflammatory signaling pathways. However, the strategy also interrupts the survival and reproduction functions conducted by TNFα-TNFR2 interaction and causes side effects. Thus, it is urgently needed to develop inhibitors that can selectively block TNFα-TNFR1 but not TNFα-TNFR2. Here, nucleic acid-based aptamers against TNFR1 are explored as potential anti-RA candidates. Through the systematic evolution of ligands by exponential enrichment (SELEX), two types of TNFR1-targeting aptamers were obtained, and their KD values are approximately 100-300 nM. In silico analysis shows that the binding interface of aptamer-TNFR1 highly overlapped with natural TNFα-TNFR1 binding. On the cellular level, the aptamers can exert TNFα inhibitory activity by binding to TNFR1. The anti-inflammatory efficiencies of aptamers were assessed and further enhanced using divalent aptamer constructs. These findings provide a new strategy to block TNFR1 for potential anti-RA treatment precisely.
Collapse
Affiliation(s)
- Xiao Chu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
| | - Xinyu Du
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
| | - Longhua Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ziyi Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yi Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaonan Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lijun Dai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jiangnan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jie Liu
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
| | - Nan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yongxing Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongzhou Gu
- Fudan University Shanghai Cancer Center, and Institutes of Biomedical Sciences, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
34
|
Tscheuschner G, Ponader M, Raab C, Weider PS, Hartfiel R, Kaufmann JO, Völzke JL, Bosc-Bierne G, Prinz C, Schwaar T, Andrle P, Bäßler H, Nguyen K, Zhu Y, Mey ASJS, Mostafa A, Bald I, Weller MG. Efficient Purification of Cowpea Chlorotic Mottle Virus by a Novel Peptide Aptamer. Viruses 2023; 15:v15030697. [PMID: 36992405 PMCID: PMC10051510 DOI: 10.3390/v15030697] [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: 01/26/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/31/2023] Open
Abstract
The cowpea chlorotic mottle virus (CCMV) is a plant virus explored as a nanotechnological platform. The robust self-assembly mechanism of its capsid protein allows for drug encapsulation and targeted delivery. Additionally, the capsid nanoparticle can be used as a programmable platform to display different molecular moieties. In view of future applications, efficient production and purification of plant viruses are key steps. In established protocols, the need for ultracentrifugation is a significant limitation due to cost, difficult scalability, and safety issues. In addition, the purity of the final virus isolate often remains unclear. Here, an advanced protocol for the purification of the CCMV from infected plant tissue was developed, focusing on efficiency, economy, and final purity. The protocol involves precipitation with PEG 8000, followed by affinity extraction using a novel peptide aptamer. The efficiency of the protocol was validated using size exclusion chromatography, MALDI-TOF mass spectrometry, reversed-phase HPLC, and sandwich immunoassay. Furthermore, it was demonstrated that the final eluate of the affinity column is of exceptional purity (98.4%) determined by HPLC and detection at 220 nm. The scale-up of our proposed method seems to be straightforward, which opens the way to the large-scale production of such nanomaterials. This highly improved protocol may facilitate the use and implementation of plant viruses as nanotechnological platforms for in vitro and in vivo applications.
Collapse
Affiliation(s)
- Georg Tscheuschner
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Marco Ponader
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Christopher Raab
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Prisca S Weider
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Reni Hartfiel
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Jan Ole Kaufmann
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, 81675 Munich, Germany
| | - Jule L Völzke
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Gaby Bosc-Bierne
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Carsten Prinz
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | | | - Paul Andrle
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Henriette Bäßler
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Khoa Nguyen
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - Yanchen Zhu
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK
| | - Antonia S J S Mey
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK
| | - Amr Mostafa
- Institute of Chemistry-Physical Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Ilko Bald
- Institute of Chemistry-Physical Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Michael G Weller
- Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| |
Collapse
|
35
|
Zhu H, Wang J, Zhang Q, Pan X, Zhang J. Novel strategies and promising opportunities for targeted protein degradation: An innovative therapeutic approach to overcome cancer resistance. Pharmacol Ther 2023; 244:108371. [PMID: 36871783 DOI: 10.1016/j.pharmthera.2023.108371] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Targeted Protein Degradation is an emerging and rapidly developing technique for designing and treating new drugs. With the emergence of a promising class of pharmaceutical molecules, Heterobifunctional Proteolysis-targeting chimeras (PROTACs), TPD has become a powerful tool to completely tackle pathogenic proteins with traditional small molecule inhibitors. However, the conventional PROTACs have gradually exposed potential disadvantages of poor oral bioavailability and pharmacokinetic (PK) and absorption, distribution, metabolism, excretion, and toxicity (ADMET) characteristics due to their larger molecular weight and more complex structure than the conventional small-molecule inhibitors. Therefore, 20 years after the concept of PROTAC was proposed, more and more scientists are committed to developing new TPD technology to overcome its defects. And several new technologies and means have been explored based on "PROTAC" to target "undruggable proteins". Here, we aim to comprehensively summarize and profoundly analyze the research progress of targeted protein degradation based on PROTAC targeting the degradation of "undruggable" targets. In order to clarify the significance of emerging and highly effective strategies based PROTACs in the treatment of various diseases especially in overcoming drug resistance in cancer, we will focus on the molecular structure, action mechanism, design concepts, development advantages and challenges of these emerging methods(e.g., aptamer-PROTAC conjugates, antibody-PROTACs and folate-PROTACs).
Collapse
Affiliation(s)
- Huanjie Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
36
|
Puumala LS, Grist SM, Morales JM, Bickford JR, Chrostowski L, Shekhar S, Cheung KC. Biofunctionalization of Multiplexed Silicon Photonic Biosensors. BIOSENSORS 2022; 13:53. [PMID: 36671887 PMCID: PMC9855810 DOI: 10.3390/bios13010053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/10/2022] [Accepted: 12/23/2022] [Indexed: 05/28/2023]
Abstract
Silicon photonic (SiP) sensors offer a promising platform for robust and low-cost decentralized diagnostics due to their high scalability, low limit of detection, and ability to integrate multiple sensors for multiplexed analyte detection. Their CMOS-compatible fabrication enables chip-scale miniaturization, high scalability, and low-cost mass production. Sensitive, specific detection with silicon photonic sensors is afforded through biofunctionalization of the sensor surface; consequently, this functionalization chemistry is inextricably linked to sensor performance. In this review, we first highlight the biofunctionalization needs for SiP biosensors, including sensitivity, specificity, cost, shelf-stability, and replicability and establish a set of performance criteria. We then benchmark biofunctionalization strategies for SiP biosensors against these criteria, organizing the review around three key aspects: bioreceptor selection, immobilization strategies, and patterning techniques. First, we evaluate bioreceptors, including antibodies, aptamers, nucleic acid probes, molecularly imprinted polymers, peptides, glycans, and lectins. We then compare adsorption, bioaffinity, and covalent chemistries for immobilizing bioreceptors on SiP surfaces. Finally, we compare biopatterning techniques for spatially controlling and multiplexing the biofunctionalization of SiP sensors, including microcontact printing, pin- and pipette-based spotting, microfluidic patterning in channels, inkjet printing, and microfluidic probes.
Collapse
Affiliation(s)
- Lauren S. Puumala
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Samantha M. Grist
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Dream Photonics Inc., Vancouver, BC V6T 0A7, Canada
| | - Jennifer M. Morales
- Army Research Laboratory, US Army Combat Capabilities Development Command, 2800 Powder Mill Rd., Adelphi, MD 20783, USA
| | - Justin R. Bickford
- Army Research Laboratory, US Army Combat Capabilities Development Command, 2800 Powder Mill Rd., Adelphi, MD 20783, USA
| | - Lukas Chrostowski
- Dream Photonics Inc., Vancouver, BC V6T 0A7, Canada
- Department of Electrical and Computer Engineering, University of British Columbia, 2332 Main Mall, Vancouver, BC V6T 1Z4, Canada
- Stewart Blusson Quantum Matter Institute, University of British Columbia, 2355 East Mall, Vancouver, BC V6T 1Z4, Canada
| | - Sudip Shekhar
- Dream Photonics Inc., Vancouver, BC V6T 0A7, Canada
- Department of Electrical and Computer Engineering, University of British Columbia, 2332 Main Mall, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Department of Electrical and Computer Engineering, University of British Columbia, 2332 Main Mall, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
37
|
Chen J, Xiang Y, Wang P, Liu J, Lai W, Xiao M, Pei H, Fan C, Li L. Ensemble Modified Aptamer Based Pattern Recognition for Adaptive Target Identification. NANO LETTERS 2022; 22:10057-10065. [PMID: 36524831 DOI: 10.1021/acs.nanolett.2c03808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The difficulty of the molecular design and chemical synthesis of artificial sensing receptors restricts their diagnostic and proteomic applications. Herein, we report a concept of "ensemble modified aptamers" (EMAmers) that exploits the collective recognition abilities of a small set of protein-like side-chain-modified nucleic acid ligands for discriminative identification of molecular or cellular targets. Different types and numbers of hydrophobic functional groups were incorporated at designated positions on nucleic acid scaffolds to mimic amino acid side chains. We successfully assayed 18 EMAmer probes with differential binding affinities to seven proteins. We constructed an EMAmer-based chemical nose sensor and demonstrated its application in blinded unknown protein identification, giving a 92.9% accuracy. Additionally, the sensor is generalizable to the detection of blinded unknown bacterial and cellular samples, which enabled identification accuracies of 96.3% and 94.8%, respectively. This sensing platform offers a discriminative means for adaptive target identification and holds great potential for diverse applications.
Collapse
Affiliation(s)
- Jing Chen
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Ying Xiang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Peipei Wang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Jingjing Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Wei Lai
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201240, People's Republic of China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, People's Republic of China
| |
Collapse
|
38
|
Oh J, Min C, Park D, Han MS. Oligonucleotide-Chemosensor Conjugate as a Dual Responsive Detection Platform and Its Application for Simultaneous Detection of ATP and Zn 2. ACS Sens 2022; 7:3933-3939. [PMID: 36503238 DOI: 10.1021/acssensors.2c02006] [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] [Indexed: 12/14/2022]
Abstract
Simultaneous detection, which helps understand complex physiological processes and accurately diagnose diseases, has been achieved using dual responsive probes. The dual responsive probe can ideally distinguish four cases, which are a combination of the absence and presence of two analytes, with characteristic fluorescence emissions. Owing to the demanding conditions of its development, most previous studies have focused on the simple linkage between small-molecule chemosensors that have an individual target and spectral range. In this study, a new dual responsive detection platform, oligonucleotide-chemosensor conjugate, was developed using a linkage between versatile oligonucleotide probes and small-molecule chemosensors to expand the applicable scaffold and detectable target for simultaneous detection. As a proof of concept, the ATP aptamer probe and Zn2+ chemosensor were conjugated as the levels of ATP and Zn2+ are intimately correlated in several signaling pathways and diseases. Each probe could detect an analyte independently within a conjugate probe, and simultaneous detection was also demonstrated without spectral crosstalk or interference between the receptors. In addition, the introduced cholesterol modification allowed the developed probe to detect changes in analytes on the plasma membrane of live cells through flow cytometry and confocal microscopy.
Collapse
Affiliation(s)
- Jinyoung Oh
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Chanhyuk Min
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Daeho Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Min Su Han
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
39
|
Shraim AS, Abdel Majeed BA, Al-Binni M, Hunaiti A. Therapeutic Potential of Aptamer-Protein Interactions. ACS Pharmacol Transl Sci 2022; 5:1211-1227. [PMID: 36524009 PMCID: PMC9745894 DOI: 10.1021/acsptsci.2c00156] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Indexed: 11/06/2022]
Abstract
Aptamers are single-stranded oligonucleotides (RNA or DNA) with a typical length between 25 and 100 nucleotides which fold into three-dimensional structures capable of binding to target molecules. Specific aptamers can be isolated against a large variety of targets through efficient and relatively cheap methods, and they demonstrate target-binding affinities that sometimes surpass those of antibodies. Consequently, interest in aptamers has surged over the past three decades, and their application has shown promise in advancing knowledge in target analysis, designing therapeutic interventions, and bioengineering. With emphasis on their therapeutic applications, aptamers are emerging as a new innovative class of therapeutic agents with promising biochemical and biological properties. Aptamers have the potential of providing a feasible alternative to antibody- and small-molecule-based therapeutics given their binding specificity, stability, low toxicity, and apparent non-immunogenicity. This Review examines the general properties of aptamers and aptamer-protein interactions that help to understand their binding characteristics and make them important therapeutic candidates.
Collapse
Affiliation(s)
- Ala’a S. Shraim
- Department
of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, 19328 Amman, Jordan
- Pharmacological
and Diagnostic Research Center (PDRC), Al-Ahliyya
Amman University, 19328 Amman, Jordan
| | - Bayan A. Abdel Majeed
- Department
of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, 19328 Amman, Jordan
- Pharmacological
and Diagnostic Research Center (PDRC), Al-Ahliyya
Amman University, 19328 Amman, Jordan
| | - Maysaa’
Adnan Al-Binni
- Department
of Clinical Laboratory Sciences, School of Science, The University of Jordan, 11942 Amman, Jordan
| | - Abdelrahim Hunaiti
- Department
of Clinical Laboratory Sciences, School of Science, The University of Jordan, 11942 Amman, Jordan
| |
Collapse
|
40
|
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
|
41
|
Zhao C, Dekker FJ. Novel Design Strategies to Enhance the Efficiency of Proteolysis Targeting Chimeras. ACS Pharmacol Transl Sci 2022; 5:710-723. [PMID: 36110375 PMCID: PMC9469497 DOI: 10.1021/acsptsci.2c00089] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Indexed: 11/30/2022]
Abstract
Despite the success of drug discovery over the past decades, many potential drug targets still remain intractable for small molecule modulation. The development of proteolysis targeting chimeras (PROTACs) that trigger degradation of the target proteins provides a conceptually novel approach to address drug targets that remained previously elusive. Currently, the main challenge of PROTAC development is the identification of efficient, tissue- and cell-selective PROTAC molecules with good drug-likeness and favorable safety profiles. This review focuses on strategies to enhance the effectiveness and selectivity of PROTACs. We provide a comprehensive summary of recently reported PROTAC design strategies and discuss the advantages and disadvantages of these strategies. Future perspectives for PROTAC design will also be discussed.
Collapse
Affiliation(s)
- Chunlong Zhao
- Department of Chemical and
Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Frank J. Dekker
- Department of Chemical and
Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands
| |
Collapse
|
42
|
Zhu C, Zhang F, Li H, Chen Z, Yan M, Li L, Qu F. CRISPR/Cas Systems Accelerating the Development of Aptasensors. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
43
|
He M, Cao C, Ni Z, Liu Y, Song P, Hao S, He Y, Sun X, Rao Y. PROTACs: great opportunities for academia and industry (an update from 2020 to 2021). Signal Transduct Target Ther 2022; 7:181. [PMID: 35680848 PMCID: PMC9178337 DOI: 10.1038/s41392-022-00999-9] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/25/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023] Open
Abstract
PROteolysis TArgeting Chimeras (PROTACs) technology is a new protein-degradation strategy that has emerged in recent years. It uses bifunctional small molecules to induce the ubiquitination and degradation of target proteins through the ubiquitin-proteasome system. PROTACs can not only be used as potential clinical treatments for diseases such as cancer, immune disorders, viral infections, and neurodegenerative diseases, but also provide unique chemical knockdown tools for biological research in a catalytic, reversible, and rapid manner. In 2019, our group published a review article "PROTACs: great opportunities for academia and industry" in the journal, summarizing the representative compounds of PROTACs reported before the end of 2019. In the past 2 years, the entire field of protein degradation has experienced rapid development, including not only a large increase in the number of research papers on protein-degradation technology but also a rapid increase in the number of small-molecule degraders that have entered the clinical and will enter the clinical stage. In addition to PROTAC and molecular glue technology, other new degradation technologies are also developing rapidly. In this article, we mainly summarize and review the representative PROTACs of related targets published in 2020-2021 to present to researchers the exciting developments in the field of protein degradation. The problems that need to be solved in this field will also be briefly introduced.
Collapse
Affiliation(s)
- Ming He
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Chaoguo Cao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
- Tsinghua-Peking Center for Life Sciences, 100084, Beijing, P. R. China
| | - Zhihao Ni
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Yongbo Liu
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Peilu Song
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Shuang Hao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Yuna He
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Xiuyun Sun
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Yu Rao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China.
- School of Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, China.
| |
Collapse
|
44
|
Yang LF, Kacherovsky N, Panpradist N, Wan R, Liang J, Zhang B, Salipante SJ, Lutz BR, Pun SH. Aptamer Sandwich Lateral Flow Assay (AptaFlow) for Antibody-Free SARS-CoV-2 Detection. Anal Chem 2022; 94:7278-7285. [PMID: 35532905 PMCID: PMC9112978 DOI: 10.1021/acs.analchem.2c00554] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/10/2022] [Indexed: 12/17/2022]
Abstract
The COVID-19 pandemic is among the greatest health and socioeconomic crises in recent history. Although COVID-19 vaccines are being distributed, there remains a need for rapid testing to limit viral spread from infected individuals. We previously identified the SARS-CoV-2 spike protein N-terminal domain (NTD) binding DNA aptamer 1 (SNAP1) for detection of SARS-CoV-2 virus by aptamer-antibody sandwich enzyme-linked immunoassay (ELISA) and lateral flow assay (LFA). In this work, we identify a new aptamer that also binds at the NTD, named SARS-CoV-2 spike protein NTD-binding DNA aptamer 4 (SNAP4). SNAP4 binds with high affinity (<30 nM) for the SARS-CoV-2 spike protein, a 2-fold improvement over SNAP1. Furthermore, we utilized both SNAP1 and SNAP4 in an aptamer sandwich LFA (AptaFlow), which detected SARS-CoV-2 UV-inactivated virus at concentrations as low as 106 copies/mL. AptaFlow costs <$1 per test to produce, provides results in <1 h, and detects SARS-CoV-2 at concentrations that indicate higher viral loads and a high probability of contagious transmission. AptaFlow is a potential approach for a low-cost, convenient antigen test to aid the control of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Lucy F. Yang
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington 98195
| | - Nataly Kacherovsky
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington 98195
| | - Nuttada Panpradist
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington 98195
| | - Ruixuan Wan
- Department of Chemistry, University of Washington, Seattle, Washington 98195
| | - Joey Liang
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington 98195
| | - Bo Zhang
- Department of Chemistry, University of Washington, Seattle, Washington 98195
| | - Stephen J. Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195
| | - Barry R. Lutz
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington 98195
| | - Suzie H. Pun
- Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, Seattle, Washington 98195
| |
Collapse
|
45
|
Chakraborty B, Das S, Gupta A, Xiong Y, Vyshnavi TV, Kizer ME, Duan J, Chandrasekaran AR, Wang X. Aptamers for Viral Detection and Inhibition. ACS Infect Dis 2022; 8:667-692. [PMID: 35220716 PMCID: PMC8905934 DOI: 10.1021/acsinfecdis.1c00546] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Indexed: 02/07/2023]
Abstract
Recent times have experienced more than ever the impact of viral infections in humans. Viral infections are known to cause diseases not only in humans but also in plants and animals. Here, we have compiled the literature review of aptamers selected and used for detection and inhibition of viral infections in all three categories: humans, animals, and plants. This review gives an in-depth introduction to aptamers, different types of aptamer selection (SELEX) methodologies, the benefits of using aptamers over commonly used antibody-based strategies, and the structural and functional mechanism of aptasensors for viral detection and therapy. The review is organized based on the different characterization and read-out tools used to detect virus-aptasensor interactions with a detailed index of existing virus-targeting aptamers. Along with addressing recent developments, we also discuss a way forward with aptamers for DNA nanotechnology-based detection and treatment of viral diseases. Overall, this review will serve as a comprehensive resource for aptamer-based strategies in viral diagnostics and treatment.
Collapse
Affiliation(s)
- Banani Chakraborty
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Sreyashi Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Arushi Gupta
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Yanyu Xiong
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory (HMNTL), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - T-V Vyshnavi
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Megan E. Kizer
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jinwei Duan
- Department of Chemistry and Materials Science, Chang’an University, Xi’an, Shaanxi 710064, China
| | - Arun Richard Chandrasekaran
- The RNA Institute, University at Albany, State University of New York, Albany, New York 12222, United States
| | - Xing Wang
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory (HMNTL), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology (IGB), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
46
|
Li Y, Song M, Gao R, Lu F, Liu J, Huang Q. Repurposing of thermally stable nucleic-acid aptamers for targeting tetrodotoxin (TTX). Comput Struct Biotechnol J 2022; 20:2134-2142. [PMID: 35832627 PMCID: PMC9092388 DOI: 10.1016/j.csbj.2022.04.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/23/2022] [Accepted: 04/23/2022] [Indexed: 01/03/2023] Open
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Centre of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Menghua Song
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Centre of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ruihua Gao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Feng Lu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Jianping Liu
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Centre of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
- Corresponding authors at: State Key Laboratory of Genetic Engineering, MOE Engineering Research Centre of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China (Q. Huang).
| | - Qiang Huang
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Centre of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 201203, China
- Corresponding authors at: State Key Laboratory of Genetic Engineering, MOE Engineering Research Centre of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China (Q. Huang).
| |
Collapse
|
47
|
Chen W, Chen Z, Lai Q, Zhang Y, Long M, Liang B, Liu Z. Specific and ultrasensitive detection of Staphylococcus aureus with a catechol-chitosan redox capacitor based electrochemical aptasensor. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2022.116357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
48
|
Nosrati M, Roushani M. Three-dimensional modeling of streptomycin binding single-stranded DNA for aptamer-based biosensors, a molecular dynamics simulation approach. J Biomol Struct Dyn 2022; 41:3430-3439. [PMID: 35297324 DOI: 10.1080/07391102.2022.2050945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Streptomycin (STR) an aminoglycoside antibiotic which is used against bacteria in human and animal infection, have serious side effects on different parts of human body. Therefore, there is a crucial need to detect trace amount of it in serum and food products. Aptamers are oligonucleotides or peptides, which bind their targets with high affinity and specificity. These properties make aptamers as suitable candidates for biosensing applications. A 79-mer ss-DNA aptamer was applied for the detection of small amount of STR in various aptasensors. But there is no structural information on the STR-binding aptamer and molecular details underlying the aptamer-STR binding remain unexplored. In this study we provided a 3D-structural model for 79-mer ss-DNA aptamer from the sequence. Using docking program and molecular dynamics (MD) simulation we predicted the binding pocket of ss-DNA aptamer. Our results show STR streptose ring is buried within the groove of DNA model and capped by non Watson-Crick bases. STR interacts with aptamer through forming stable hydrogen bonds. Our computational findings are in fair agreement with experimental results. With the atomic structural details, we gained new insight into the Apt-STR binding interaction that can help to further optimize aptamer efficiency in biosensing applications.Communicated by Ramaswamy H. Sarma.
Collapse
|
49
|
Bao Y, Zhu D, Zhao Y, Li X, Gu C, Yu H. Selection and identification of high-affinity aptamer of Kunitz trypsin inhibitor and their application in rapid and specific detection. Food Sci Nutr 2022; 10:953-963. [PMID: 35282009 PMCID: PMC8907715 DOI: 10.1002/fsn3.2729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/30/2021] [Accepted: 01/02/2022] [Indexed: 12/21/2022] Open
Abstract
Kunitz trypsin inhibitor (KTI), a harmful protein, seriously affects food hygiene and safety. Therefore, a sensitive, efficient, and rapid method for KTI detection is urgently needed. Aptamers are short and single-stranded (ss) DNA that recognize target molecules with high affinity. This work used graphene oxide-SELEX (GO-SELEX) to screen KTI aptamers. The positive and reverse screening was designed to ensure the high specificity and affinity of the selected aptamers. After 10 rounds of screening, multiple nucleic acid chains were obtained, and the chains were sequenced. Three aptamers with better affinity were obtained, and the values of the dissociation constant (K d) were calculated to be 52.6 nM, 22.7 nM, and 67.9 nM, respectively. Finally, a colorimetric aptamer biosensor based on gold nanoparticles (AuNPs) was constructed. The biosensor exhibited a broader linear range of 30-750 ng/ml, with a lower detection limit of 18 ng/ml, and the spiked recovery rate was between 98.2% and 103.3%. This experiment preliminary demonstrated the potential of the application of KTI aptamer in the real sample tests.
Collapse
Affiliation(s)
- Yunxiang Bao
- College of Food Science and EngineeringJilin Agricultural UniversityChangchunChina
| | - Dengzhao Zhu
- College of Food Science and EngineeringJilin Agricultural UniversityChangchunChina
| | - Yang Zhao
- College of Food Science and EngineeringJilin Agricultural UniversityChangchunChina
- Division of Soybean ProcessingSoybean Research & Development CenterChinese Agricultural Research SystemChangchunChina
| | - Xinzhu Li
- College of Food Science and EngineeringJilin Agricultural UniversityChangchunChina
| | - Chunmei Gu
- College of Food Science and EngineeringJilin Agricultural UniversityChangchunChina
| | - Hansong Yu
- College of Food Science and EngineeringJilin Agricultural UniversityChangchunChina
- Division of Soybean ProcessingSoybean Research & Development CenterChinese Agricultural Research SystemChangchunChina
| |
Collapse
|
50
|
Xia F, He A, Zhao H, Sun Y, Duan Q, Abbas SJ, Liu J, Xiao Z, Tan W. Molecular Engineering of Aptamer Self-Assemblies Increases in Vivo Stability and Targeted Recognition. ACS NANO 2022; 16:169-179. [PMID: 34935348 DOI: 10.1021/acsnano.1c05265] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Functionally modified aptamer conjugates are promising tools for targeted imaging or treatment of various diseases. However, broad applications of aptamer molecules are limited by their in vivo instability. To overcome this challenge, current strategies mostly rely on covalent chemical modification of aptamers, a complicated process that requires case-by-case sequence design, multiple-step synthesis, and purification. Herein, we report a covalent modification-free strategy to enhance the in vivo stability of aptamers. This strategy simply utilizes one-step molecular engineering of aptamers with gold nanoclusters (GNCs) to form GNCs@aptamer self-assemblies. Using Sgc8 as a representative aptamer, the resulting GNCs@Sgc8 assemblies enhance cancer-cell-specific binding and sequential internalization by a receptor-mediated endocytosis pathway. Importantly, the GNCs@aptamer self-assemblies resist nuclease degradation for as long as 48 h, compared to the degradation of aptamer alone at 3 h. In parallel, the tumor-targeted recognition and retention of GNCs@aptamer self-assemblies are dramatically enhanced, indicated by a 9-fold signal increase inside the tumor compared to the aptamer alone. This strategy is to avoid complicated chemical modification of aptamers and can be extended to all aptamers. Our work provides a simple, effective, and universal strategy for enhancing the in vivo stability of any aptamer or its conjugates, thus expanding their imaging and therapeutic applications.
Collapse
Affiliation(s)
- Fangfang Xia
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Axin He
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haitao Zhao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yang Sun
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiao Duan
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Sk Jahir Abbas
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zeyu Xiao
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Pharmacology and Chemical Biology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), 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
| |
Collapse
|