1
|
Zhou P, Zhang S, Li L, Zhang R, Guo G, Zhang Y, Wang R, Liu M, Wang Z, Zhao H, Yang G, Xie S, Ran J. Targeted degradation of VEGF with bispecific aptamer-based LYTACs ameliorates pathological retinal angiogenesis. Theranostics 2024; 14:4983-5000. [PMID: 39267779 PMCID: PMC11388081 DOI: 10.7150/thno.98467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024] Open
Abstract
Rationale: Neovascular ocular diseases (NODs) represent the leading cause of visual impairment globally. Despite significant advances in anti-angiogenic therapies targeting vascular endothelial growth factor (VEGF), persistent challenges remain prevalent. As a proof-of-concept study, we herein demonstrate the effectiveness of targeted degradation of VEGF with bispecific aptamer-based lysosome-targeting chimeras (referred to as VED-LYTACs). Methods: VED-LYTACs were constructed with three distinct modules: a mannose-6-phosphate receptor (M6PR)-binding motif containing an M6PR aptamer, a VEGF-binding module with an aptamer targeting VEGF, and a linker essential for bridging and stabilizing the two-aptamer structure. The degradation efficiency of VED-LYTACs via the autophagy-lysosome system was examined using an enzyme-linked immunosorbent assay (ELISA) and immunofluorescence staining. Subsequently, the anti-angiogenic effects of VED-LYTACs were evaluated using in vitro wound healing assay, tube formation assay, three-dimensional sprouting assay, and ex vivo aortic ring sprouting assay. Finally, the potential therapeutic effects of VED-LYTACs on pathological retinal neovascularization and vascular leakage were tested by employing mouse models of NODs. Results: The engineered VED-LYTACs promote the interaction between M6PR and VEGF, consequently facilitating the translocation and degradation of VEGF through the lysosome. Our data show that treatment with VED-LYTACs significantly suppresses VEGF-induced angiogenic activities both in vitro and ex vivo. In addition, intravitreal injection of VED-LYTACs remarkably ameliorates abnormal vascular proliferation and leakage in mouse models of NODs. Conclusion: Our findings present a novel strategy for targeting VEGF degradation with an aptamer-based LYTAC system, effectively ameliorating pathological retinal angiogenesis. These results suggest that VED-LYTACs have potential as therapeutic agents for managing NODs.
Collapse
Affiliation(s)
- Ping Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Sai Zhang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Lin Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Renshuai Zhang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Guizhi Guo
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yufei Zhang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Runa Wang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Miaoyuan Liu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Zhiyi Wang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Guiwen Yang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Songbo Xie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
2
|
Go YJ, Kalathingal M, Rhee YM. An Ensemble Docking Approach for Analyzing and Designing Aptamer Heterodimers Targeting VEGF 165. Int J Mol Sci 2024; 25:4066. [PMID: 38612876 PMCID: PMC11012306 DOI: 10.3390/ijms25074066] [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: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Vascular endothelial growth factor 165 (VEGF165) is a prominent isoform of the VEGF-A protein that plays a crucial role in various angiogenesis-related diseases. It is homodimeric, and each of its monomers is composed of two domains connected by a flexible linker. DNA aptamers, which have emerged as potent therapeutic molecules for many proteins with high specificity and affinity, can also work for VEGF165. A DNA aptamer heterodimer composed of monomers of V7t1 and del5-1 connected by a flexible linker (V7t1:del5-1) exhibits a greater binding affinity with VEGF165 compared to either of the two monomers alone. Although the structure of the complex formed between the aptamer heterodimer and VEGF165 is unknown due to the highly flexible linkers, gaining structural information will still be valuable for future developments. Toward this end of accessing structural information, we adopt an ensemble docking approach here. We first obtain an ensemble of structures for both VEGF165 and the aptamer heterodimer by considering both small- and large-scale motions. We then proceed through an extraction process based on ensemble docking, molecular dynamics simulations, and binding free energy calculations to predict the structures of the VEGF165/V7t1:del5-1 complex. Through the same procedures, we reach a new aptamer heterodimer that bears a locked nucleic acid-modified counterpart of V7t1, namely RNV66:del5-1, which also binds well with VEGF165. We apply the same protocol to the monomeric units V7t1, RNV66, and del5-1 to target VEGF165. We observe that V7t1:del5-1 and RNV66:del5-1 show higher binding affinities with VEGF165 than any of the monomers, consistent with experiments that support the notion that aptamer heterodimers are more effective anti-VEGF165 aptamers than monomeric aptamers. Among the five different aptamers studied here, the newly designed RNV66:del5-1 shows the highest binding affinity with VEGF165. We expect that our ensemble docking approach can help in de novo designs of homo/heterodimeric anti-angiogenic drugs to target the homodimeric VEGF165.
Collapse
Affiliation(s)
- Yeon Ju Go
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea;
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Mahroof Kalathingal
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea;
| | - Young Min Rhee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea;
| |
Collapse
|
3
|
Sriondee Y, Vijitvarasan P, Rattanachata A, Nakajima H, Oaew S, Cheunkar S. Real-time kinetic analysis and detection of glycated hemoglobin A1c using a quartz crystal microbalance-based aptasensor. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:599-607. [PMID: 38197200 DOI: 10.1039/d3ay01842c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Glycated hemoglobin (HbA1c) has been an important biomarker for long-term diagnosis and monitoring of diabetes mellitus. The development of a rapid, reliable, and less sophisticated device to measure HbA1c is imperative to facilitate efficient early-care diabetes management. To date, no existing aptamer-based biosensor (aptasensor) for detecting HbA1c has been developed using a quartz crystal microbalance (QCM). In this study, the aptamer specific to HbA1c as a novel biosensing receptor was covalently functionalized onto a QCM substrate via mixed self-assembled monolayers (SAMs). A portable QCM equipped with a liquid-flow module was used to investigate the biospecificity, sensitivity, and interaction dynamics of the aptamer functionalized surfaces. The real-time kinetic analysis of HbA1c binding to the surface-functionalized aptamers revealed "on" and "off" binding rates of 4.19 × 104 M-1 s-1 and 2.43 × 10-3 s-1, respectively. These kinetic parameters imply that the QCM-based aptasensor specifically recognizes HbA1c with an equilibrium dissociation constant as low as 57.99 nM. The linear detection of HbA1c spanned from 13 to 108 nM, with a limit of detection (LOD) of 26.29 nM. Moreover, the spiked plasma sample analysis offered compelling evidence that this aptasensor is a promising technique for developing a point-of-care device for diabetes mellitus.
Collapse
Affiliation(s)
- Yossawadee Sriondee
- Division of Biotechnology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand.
| | | | | | - Hideki Nakajima
- Synchrotron Light Research Institute, Nakhon Ratchasima, 30000, Thailand
| | - Sukunya Oaew
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand.
| | - Sarawut Cheunkar
- Division of Biotechnology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand.
| |
Collapse
|
4
|
Cai D, Chen GL, Wang T, Zhang KH. Combination of multiple nucleic acid aptamers for precision detection of tumors based on optical methods. J Cancer Res Clin Oncol 2023; 149:7895-7903. [PMID: 36809501 DOI: 10.1007/s00432-023-04646-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 02/09/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND PURPOSE Nucleic acid aptamers are a novel molecular recognition tool that is functionally similar to antibodies but superior to antibodies in terms of thermal stability, structural modification, preparation, and cost, and therefore hold great promise for molecular detection. However, due to the limitations of a single aptamer in molecular detection, the multiple aptamer combination for bioanalysis has received much attention. Here, we reviewed the progress of tumor precision detection based on the combination of multiple nucleic acid aptamers and optical methods and discussed its challenges and prospects. METHODS The relevant literature in PubMed was collected and reviewed. RESULTS The combination of two or more aptamers with modern nanomaterials and analytical methods allows the fabrication of various detection systems for the simultaneous detection of different structural domains of a substance and/or different substances, including soluble tumor markers, tumor cell surface and intracellular markers, circulating tumor cells, and other tumor-related biomolecules, which has great potential for application in efficient and precise tumor detection. CONCLUSION The combination of multiple nucleic acid aptamers provides a new approach for the precise detection of tumors and will play an important role in precision medicine for tumors.
Collapse
Affiliation(s)
- Dan Cai
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Gastroenterology and Hepatology, No 17, Yongwai Zheng Street, Nanchang, 330006, Jiangxi, China
| | - Gui-Lin Chen
- Department of Anorectal Surgery, Chinese People's Liberation Army Joint Security Force Hospital No. 908, No 1028, Jinggangshan Avenue, Nanchang, China
| | - Ting Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Gastroenterology and Hepatology, No 17, Yongwai Zheng Street, Nanchang, 330006, Jiangxi, China
| | - Kun-He Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Institute of Gastroenterology and Hepatology, No 17, Yongwai Zheng Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
5
|
Díaz-Fernández A, Ferapontov A, Vendelbo MH, Ferapontova EE. Electrochemical Cellulase-Linked ELASA for Rapid Liquid Biopsy Testing of Serum HER-2/ neu. ACS MEASUREMENT SCIENCE AU 2023; 3:226-235. [PMID: 37360033 PMCID: PMC10288612 DOI: 10.1021/acsmeasuresciau.2c00067] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 06/28/2023]
Abstract
Non-invasive liquid biopsy assays for blood-circulating biomarkers of cancer allow both its early diagnosis and treatment monitoring. Here, we assessed serum levels of protein HER-2/neu, overexpressed in a number of aggressive cancers, by the cellulase-linked sandwich bioassay on magnetic beads. Instead of traditional antibodies we used inexpensive reporter and capture aptamer sequences, transforming the enzyme-linked immuno-sorbent assay (ELISA) into an enzyme-linked aptamer-sorbent assay (ELASA). The reporter aptamer was conjugated to cellulase, whose digestion of nitrocellulose film electrodes resulted in the electrochemical signal change. ELASA, optimized relative aptamer lengths (dimer vs monomer and trimer), and assay steps allowed 0.1 fM detection of HER-2/neu in the 10% human serum in 1.3 h. Urokinase plasminogen activator and thrombin as well as human serum albumin did not interfere, and liquid biopsy analysis of serum HER-2/neu was similarly robust but 4 times faster and 300 times cheaper than both electrochemical and optical ELISA. Simplicity and low cost of cellulase-linked ELASA makes it a perspective diagnostic tool for fast and accurate liquid biopsy detection of HER-2/neu and of other proteins for which aptamers are available.
Collapse
Affiliation(s)
- Ana Díaz-Fernández
- Interdisciplinary
Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Alexey Ferapontov
- Interdisciplinary
Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Mikkel Holm Vendelbo
- Department
of Nuclear Medicine & PET Centre, Aarhus
University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
- Department
of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Elena E. Ferapontova
- Interdisciplinary
Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| |
Collapse
|
6
|
Torabi M, Aghanejad A, Savadi P, Barzegari A, Omidi Y, Barar J. Targeted Delivery of Sunitinib by MUC-1 Aptamer-Capped Magnetic Mesoporous Silica Nanoparticles. Molecules 2023; 28:molecules28010411. [PMID: 36615606 PMCID: PMC9824472 DOI: 10.3390/molecules28010411] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Magnetic mesoporous silica nanoparticles (MMSNPs) are being widely investigated as multifunctional novel drug delivery systems (DDSs) and play an important role in targeted therapy. Here, magnetic cores were synthesized using the thermal decomposition method. Further, to improve the biocompatibility and pharmacokinetic behavior, mesoporous silica was synthesized using the sol-gel process to coat the magnetic cores. Subsequently, sunitinib (SUN) was loaded into the MMSNPs, and the particles were armed with amine-modified mucin 1 (MUC-1) aptamers. The MMSNPs were characterized using FT-IR, TEM, SEM, electrophoresis gel, DLS, and EDX. MTT assay, flow cytometry analysis, ROS assessment, and mitochondrial membrane potential analysis evaluated the nanoparticles' biological impacts. The physicochemical analysis revealed that the engineered MMSNPs have a smooth surface and spherical shape with an average size of 97.6 nm. The biological in vitro analysis confirmed the highest impacts of the targeted MMSNPs in MUC-1 overexpressing cells (OVCAR-3) compared to the MUC-1 negative MDA-MB-231 cells. In conclusion, the synthesized MMSNP-SUN-MUC-1 nanosystem serves as a unique multifunctional targeted delivery system to combat the MUC-1 overexpressing ovarian cancer cells.
Collapse
Affiliation(s)
- Mitra Torabi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 516664-14766, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Correspondence: (A.A.); or (J.B.); Tel./Fax: +98-41-33367929 (A.A.); +1-(954)-262-1878 (J.B.)
| | - Pouria Savadi
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (Di.S.T.A.Bi.F.), University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Jaleh Barar
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 516664-14766, Iran
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
- Correspondence: (A.A.); or (J.B.); Tel./Fax: +98-41-33367929 (A.A.); +1-(954)-262-1878 (J.B.)
| |
Collapse
|
7
|
Li J, Zhang Z, Gu J, Amini R, Mansfield AG, Xia J, White D, Stacey HD, Ang JC, Panesar G, Capretta A, Filipe CDM, Mossman K, Salena BJ, Gubbay JB, Balion C, Soleymani L, Miller MS, Yamamura D, Brennan JD, Li Y. Three on Three: Universal and High-Affinity Molecular Recognition of the Symmetric Homotrimeric Spike Protein of SARS-CoV-2 with a Symmetric Homotrimeric Aptamer. J Am Chem Soc 2022; 144:23465-23473. [PMID: 36520671 PMCID: PMC9762500 DOI: 10.1021/jacs.2c09870] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Indexed: 12/23/2022]
Abstract
Our previously discovered monomeric aptamer for SARS-CoV-2 (MSA52) possesses a universal affinity for COVID-19 spike protein variants but is ultimately limited by its ability to bind only one subunit of the spike protein. The symmetrical shape of the homotrimeric SARS-CoV-2 spike protein presents the opportunity to create a matching homotrimeric molecular recognition element that is perfectly complementary to its structural scaffold, causing enhanced binding affinity. Here, we describe a branched homotrimeric aptamer with three-fold rotational symmetry, named TMSA52, that not only possesses excellent binding affinity but is also capable of binding several SARS-CoV-2 spike protein variants with picomolar affinity, as well as pseudotyped lentiviruses expressing SARS-CoV-2 spike protein variants with femtomolar affinity. Using Pd-Ir nanocubes as nanozymes in an enzyme-linked aptamer binding assay (ELABA), TMSA52 was capable of sensitively detecting diverse pseudotyped lentiviruses in pooled human saliva with a limit of detection as low as 6.3 × 103 copies/mL. The ELABA was also used to test 50 SARS-CoV-2-positive and 60 SARS-CoV-2-negative patient saliva samples, providing sensitivity and specificity values of 84.0 and 98.3%, respectively, thus highlighting the potential of TMSA52 for the development of future rapid tests.
Collapse
Affiliation(s)
- Jiuxing Li
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Zijie Zhang
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Jimmy Gu
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Ryan Amini
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Alexandria G. Mansfield
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Jianrun Xia
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Dawn White
- Biointerfaces
Institute, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4O3, Canada
| | - Hannah D. Stacey
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Michael
G. DeGroote Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- McMaster
Immunology Research Centre, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Jann C. Ang
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Michael
G. DeGroote Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- McMaster
Immunology Research Centre, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Gurpreet Panesar
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Alfredo Capretta
- Biointerfaces
Institute, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4O3, Canada
- Michael
G. DeGroote Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Carlos D. M. Filipe
- Department
of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Karen Mossman
- McMaster
Immunology Research Centre, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Department
of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Bruno J. Salena
- Department
of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | | | - Cynthia Balion
- Department
of Pathology and Molecular Medicine, McMaster
University, 1280 Main
Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Leyla Soleymani
- Michael
G. DeGroote Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Department
of Engineering Physics, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- School
of Biomedical Engineering, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Matthew S. Miller
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Michael
G. DeGroote Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- McMaster
Immunology Research Centre, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Deborah Yamamura
- Michael
G. DeGroote Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Department
of Pathology and Molecular Medicine, McMaster
University, 1280 Main
Street West, Hamilton, Ontario L8S 4K1, Canada
| | - John D. Brennan
- Biointerfaces
Institute, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4O3, Canada
| | - Yingfu Li
- Department
of Biochemistry and Biomedical Sciences, McMaster University,1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- Biointerfaces
Institute, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4O3, Canada
- Michael
G. DeGroote Institute of Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- School
of Biomedical Engineering, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
8
|
Amini R, Zhang Z, Li J, Gu J, Brennan JD, Li Y. Aptamers for SARS-CoV-2: Isolation, Characterization, and Diagnostic and Therapeutic Developments. ANALYSIS & SENSING 2022; 2:e202200012. [PMID: 35574520 PMCID: PMC9082509 DOI: 10.1002/anse.202200012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/19/2022] [Indexed: 12/17/2022]
Abstract
The SARS-CoV-2 virus and COVID-19 pandemic continue to demand effective diagnostic and therapeutic solutions. Finding these solutions requires highly functional molecular recognition elements. Nucleic acid aptamers represent a possible solution. Characterized by their high affinity and specificity, aptamers can be rapidly identified from random-sequence nucleic acid libraries. Over the past two years, many labs around the world have rushed to create diverse aptamers that target two important structural proteins of SARS-CoV-2: the spike (S) protein and nucleocapsid (N) protein. These have led to the identification of many aptamers that show real promise for the development of diagnostic tests and therapeutic agents for SARS-CoV-2. Herein we review all these developments, with a special focus on the development of diverse aptasensors for detecting SARS-CoV-2. These include electrochemical and optical sensors, lateral flow devices, and aptamer-linked immobilized sorbent assays.
Collapse
Affiliation(s)
- Ryan Amini
- Department of Biochemistry and Biomedical SciencesMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Zijie Zhang
- Department of Biochemistry and Biomedical SciencesMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Jiuxing Li
- Department of Biochemistry and Biomedical SciencesMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Jimmy Gu
- Department of Biochemistry and Biomedical SciencesMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - John D. Brennan
- Biointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical SciencesMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
- Biointerfaces InstituteMcMaster University1280 Main Street WestHamiltonOntarioL8S 4K1Canada
| |
Collapse
|
9
|
Tang T, Liu Y, Jiang Y. Recent Progress on Highly Selective and Sensitive Electrochemical Aptamer-based Sensors. Chem Res Chin Univ 2022; 38:866-878. [PMID: 35530120 PMCID: PMC9069955 DOI: 10.1007/s40242-022-2084-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/10/2022] [Indexed: 12/31/2022]
Abstract
Highly selective, sensitive, and stable biosensors are essential for the molecular level understanding of many physiological activities and diseases. Electrochemical aptamer-based (E-AB) sensor is an appealing platform for measurement in biological system, attributing to the combined advantages of high selectivity of the aptamer and high sensitivity of electrochemical analysis. This review summarizes the latest development of E-AB sensors, focuses on the modification strategies used in the fabrication of sensors and the sensing strategies for analytes of different sizes in biological system, and then looks forward to the challenges and prospects of the future development of electrochemical aptamer-based sensors.
Collapse
Affiliation(s)
- Tianwei Tang
- College of Chemistry, Beijing Normal University, Beijing, 100875 P. R. China
| | - Yinghuan Liu
- College of Chemistry, Beijing Normal University, Beijing, 100875 P. R. China
| | - Ying Jiang
- College of Chemistry, Beijing Normal University, Beijing, 100875 P. R. China
- Beijing National Laboratory for Molecular Sciences, Beijing, 100190 P. R. China
| |
Collapse
|
10
|
Assays to Estimate the Binding Affinity of Aptamers. Talanta 2022; 238:122971. [PMID: 34857318 DOI: 10.1016/j.talanta.2021.122971] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023]
Abstract
Aptamers have become coming-of-age molecular recognition elements in both diagnostic and therapeutic applications. Generated by SELEX, the 'quality control' of aptamers, which involves the validation of their binding affinity against their respective targets is pivotal to ascertain their potency prior to use in any downstream assays or applications. Several aptamers have been isolated thus far, however, the usage of inappropriate validation assays renders some of these aptamers dubitable in terms of their binding capabilities. Driven by this need, we provide an up-to-date critical review of the various strategies used to determine the aptamer-target binding affinity with the aim of providing researchers a better comprehension of the different analytical approaches in respect to the molecular properties of aptamers and their intended targets. The techniques reported have been classified as label-based techniques such as fluorescence intensity, fluorescence anisotropy, filter-binding assays, gel shift assays, ELISA; and label-free techniques such as UV-Vis spectroscopy, circular dichroism, isothermal titration calorimetry, native electrospray ionization-mass spectrometry, quartz crystal microbalance, surface plasmon resonance, NECEEM, backscattering interferometry, capillary electrophoresis, HPLC, and nanoparticle aggregation assays. Hybrid strategies combining the characteristics of both categories such as microscale thermophoresis have been also additionally emphasized. The fundamental principles, complexity, benefits, and challenges under each technique are elaborated in detail.
Collapse
|
11
|
Daniels J, Wadekar S, DeCubellis K, Jackson GW, Chiu AS, Pagneux Q, Saada H, Engelmann I, Ogiez J, Loze-Warot D, Boukherroub R, Szunerits S. A mask-based diagnostic platform for point-of-care screening of Covid-19. Biosens Bioelectron 2021; 192:113486. [PMID: 34260968 PMCID: PMC8264268 DOI: 10.1016/j.bios.2021.113486] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/02/2021] [Indexed: 12/29/2022]
Abstract
Diagnostics of SARS-CoV-2 infection using real-time reverse-transcription polymerase chain reaction (RT-PCR) on nasopharyngeal swabs is now well-established, with saliva-based testing being lately more widely implemented for being more adapted for self-testing approaches. In this study, we introduce a different concept based on exhaled breath condensate (EBC), readily collected by a mask-based sampling device, and detection with an electrochemical biosensor with a modular architecture that enables fast and specific detection and quantification of COVID-19. The face mask forms an exhaled breath vapor containment volume to hold the exhaled breath vapor in proximity to the EBC collector to enable a condensate-forming surface, cooled by a thermal mass, to coalesce the exhaled breath into a 200-500 μL fluid sample in 2 min. EBC RT-PCR for SARS-CoV-2 genes (E, ORF1ab) on samples collected from 7 SARS-CoV-2 positive and 7 SARS-CoV-2 negative patients were performed. The presence of SARS-CoV-2 could be detected in 5 out of 7 SARS-CoV-2 positive patients. Furthermore, the EBC samples were screened on an electrochemical aptamer biosensor, which detects SARS-CoV-2 viral particles down to 10 pfu mL-1 in cultured SARS-CoV-2 suspensions. Using a "turn off" assay via ferrocenemethanol redox mediator, results about the infectivity state of the patient are obtained in 10 min.
Collapse
Affiliation(s)
- John Daniels
- Diagmetrics Inc., 30 Renees Way Madison, Connecticut, 06443, USA
| | - Shekhar Wadekar
- Diagmetrics Inc., 30 Renees Way Madison, Connecticut, 06443, USA
| | - Ken DeCubellis
- Diagmetrics Inc., 30 Renees Way Madison, Connecticut, 06443, USA
| | - George W Jackson
- Base Pair Biotechnologies, Inc., 8619 Broadway St., Suite 100, Pearland, TX, USA
| | - Alexander S Chiu
- Base Pair Biotechnologies, Inc., 8619 Broadway St., Suite 100, Pearland, TX, USA
| | - Quentin Pagneux
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Hiba Saada
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Ilka Engelmann
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Judith Ogiez
- Univ Lille, CHU Lille, Laboratoire de Virologie ULR3610, F-59000, Lille, France
| | - Delphine Loze-Warot
- CerbaHealthCare Biomedical Laboratory, CERBALLIANCE Lille, 17/24 Rue de La Digue, 59000, Lille, France
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Sabine Szunerits
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France.
| |
Collapse
|
12
|
Kim W, Park J, Kim W, Jo S, Kim M, Kim C, Park H, Bang D, Lee W, Park J. Bio-inspired Ag nanovilli-based sandwich-type SERS aptasensor for ultrasensitive and selective detection of 25-hydroxy vitamin D 3. Biosens Bioelectron 2021; 188:113341. [PMID: 34044348 DOI: 10.1016/j.bios.2021.113341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023]
Abstract
Vitamin D has been identified as an essential biomarker for various diseases such as rheumatoid arthritis, cancer, and cardiovascular diseases. Recently, many reports have demonstrated a potential link between vitamin D and systemic infections, including coronavirus disease 2019. The villi of the small intestine increase the surface area of the intestinal walls, demonstrating exceptionally efficient absorption of nutrients in the lumen and adding digestive secretions. In this study, based on the villi structure, we developed a bio-inspired silver nanovilli-based sandwich-type surface enhanced Raman scattering aptasensor for the ultrasensitive and selective detection of 25-hydroxy vitamin D3. The densely packed nanovilli structure enhanced the Raman signal, forming hotspots owing to its large surface area. Using experiments and electromagnetic simulations, we optimized the nanovilli structure as a SERS sensor. The sandwich-type aptasensor was designed using an aptamer and 4-Phenyl-1,2,4-triazoline-3,5-dione-methylene blue complex. The nanovilli-based aptasensor could sensitively detect various concentrations of 25-hydroxy vitamin D3, ranging from those found in deficient to excess conditions. The detection limit of the nanovilli-based sandwich-type aptasensor for 25-hydroxy vitamin D3 was 0.001 ng/mL, which is much lower than the deficiency concentration, and was detectable even in the human serum. In addition, our proposed sensor exhibited good repeatability (17.76%) and reproducibility (7.47%). Moreover, the nanovilli-based sandwich-type SERS aptasensor could selectively distinguish 25-hydroxy vitamin D3 from other vitamins. The silver nanovilli-based sandwich-type surface enhanced Raman scattering aptasensor opens a new avenue for the development of a bio-inspired vitamin-sensing platform.
Collapse
Affiliation(s)
- Woochang Kim
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea
| | - Joohyung Park
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea
| | - Woong Kim
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea
| | - Seongjae Jo
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea; Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, South Korea
| | - Minwoo Kim
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea
| | - Chihyun Kim
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea
| | - Hyunjun Park
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea
| | - Doyeon Bang
- College of AI Convergence, Chonnam National University, Gwangju, 61186, South Korea; Korea Institute of Medical Microrobotics, Gwangju, 61011, South Korea
| | - Wonseok Lee
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea; Department of Electrical Engineering, Korea National University of Transportation, Chungju, 27469, South Korea.
| | - Jinsung Park
- Department of Control and Instrumentation Engineering, Korea University, Sejong, 30019, South Korea.
| |
Collapse
|
13
|
Citartan M. Aptamers as the powerhouse of dot blot assays. Talanta 2021; 232:122436. [PMID: 34074421 DOI: 10.1016/j.talanta.2021.122436] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022]
Abstract
Dot blot assays have always been associated with antibodies as the main molecular recognition element, which are widely employed in a myriad of diagnostic applications. With the rising of aptamers as the equivalent molecular recognition elements of antibodies, dot blot assays are also one of the diagnostic avenues that should be scrutinized for their amenability with aptamers as the potential surrogates of antibodies. In this review, the stepwise procedures of an aptamer-based dot blot assays are underscored before reviewing the existing aptamer-based dot blot assays developed so far. Most of the applications center on monitoring the progress of SELEX and as the validatory assays to assess the potency of aptamer candidates. For the purpose of diagnostics, the current effort is still languid and as such possible suggestions to galvanize the move to spur the aptamer-based dot blot assays to a point-of-care arena are discussed.
Collapse
Affiliation(s)
- Marimuthu Citartan
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia.
| |
Collapse
|
14
|
Wang T, Chen L, Chikkanna A, Chen S, Brusius I, Sbuh N, Veedu RN. Development of nucleic acid aptamer-based lateral flow assays: A robust platform for cost-effective point-of-care diagnosis. Theranostics 2021; 11:5174-5196. [PMID: 33859741 PMCID: PMC8039946 DOI: 10.7150/thno.56471] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Lateral flow assay (LFA) has made a paradigm shift in the in vitro diagnosis field due to its rapid turnaround time, ease of operation and exceptional affordability. Currently used LFAs predominantly use antibodies. However, the high inter-batch variations, error margin and storage requirements of the conventional antibody-based LFAs significantly impede its applications. The recent progress in aptamer technology provides an opportunity to combine the potential of aptamer and LFA towards building a promising platform for highly efficient point-of-care device development. Over the past decades, different forms of aptamer-based LFAs have been introduced for broad applications ranging from disease diagnosis, agricultural industry to environmental sciences, especially for the detection of antibody-inaccessible small molecules such as toxins and heavy metals. But commercial aptamer-based LFAs are still not used widely compared with antibodies. In this work, by analysing the key issues of aptamer-based LFA design, including immobilization strategies, signalling methods, and target capturing approaches, we provide a comprehensive overview about aptamer-based LFA design strategies to facilitate researchers to develop optimised aptamer-based LFAs.
Collapse
Affiliation(s)
- Tao Wang
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
- Perron Institute for Neurological and Translational Science, Perth 6009, Australia
| | - Lanmei Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
- Guangdong Key Laboratory for Research and Development of Nature Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Arpitha Chikkanna
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Suxiang Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Isabell Brusius
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Nabayet Sbuh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Rakesh N. Veedu
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
- Perron Institute for Neurological and Translational Science, Perth 6009, Australia
| |
Collapse
|
15
|
Man J, Dong J, Wang Y, He L, Yu S, Yu F, Wang J, Tian Y, Liu L, Han R, Guo H, Wu Y, Qu L. Simultaneous Detection of VEGF and CEA by Time-Resolved Chemiluminescence Enzyme-Linked Aptamer Assay. Int J Nanomedicine 2020; 15:9975-9985. [PMID: 33363367 PMCID: PMC7754089 DOI: 10.2147/ijn.s286317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As two important tumor markers, vascular endothelial growth factor (VEGF) and carcinoembryonic antigen (CEA) have a great value for clinical application in the early diagnosis of cancer. Due to the complex composition of biological samples, the results from combined detection of CEA and VEGF are often taken as a comprehensive indicator in order to make an accurate judgment on a disease. However, most of the current methods can only be used to detect the content of one biomarker. Therefore, it is necessary to explore a simple, rapid, low-cost, and highly sensitive method for the simultaneous detection of CEA and VEGF. METHODS Based on specific aptamers and magnetic separation, a time-resolved chemiluminescence enzyme-linked aptamer assay was developed for the simultaneous detections of CEA and VEGF in serum samples. RESULTS Under the optimal conditions, the linear range of the calibration curve for VEGF was from 0.5 to 80 ng mL-1, and the limit of detection was 0.1 ng mL-1. The linear range of the calibration curve for CEA was 0.5 to 160 ng mL-1, and the limit of detection was 0.1 ng mL-1. The established method was applied to detect VEGF and CEA in serum samples. The results were consistent with those of commercial kits. CONCLUSION The method has high sensitivity and can quickly obtain accurate results, which could greatly improve the measurement efficiency, reduce the cost, and also reduce the volume of sample consumed. It can be seen that the method established in this study has important application value and broad application prospect in clinical diagnosis.
Collapse
Affiliation(s)
- Jin Man
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Jiajia Dong
- College of Chemistry, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Yilin Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Leiliang He
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Songcheng Yu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Fei Yu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Jia Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Yongmei Tian
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Lie Liu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Runping Han
- College of Chemistry, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Hongchao Guo
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
- The Key Laboratory of Nanomedicine and Health Inspection of Zhengzhou, Zhengzhou, Henan450001, People’s Republic of China
| | - Lingbo Qu
- College of Chemistry, Zhengzhou University, Zhengzhou, Henan450001, People’s Republic of China
- Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou, Henan450001, People’s Republic of China
| |
Collapse
|
16
|
Riccardi C, Napolitano E, Musumeci D, Montesarchio D. Dimeric and Multimeric DNA Aptamers for Highly Effective Protein Recognition. Molecules 2020; 25:E5227. [PMID: 33182593 PMCID: PMC7698228 DOI: 10.3390/molecules25225227] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/14/2022] Open
Abstract
Multivalent interactions frequently occur in biological systems and typically provide higher binding affinity and selectivity in target recognition than when only monovalent interactions are operative. Thus, taking inspiration by nature, bivalent or multivalent nucleic acid aptamers recognizing a specific biological target have been extensively studied in the last decades. Indeed, oligonucleotide-based aptamers are suitable building blocks for the development of highly efficient multivalent systems since they can be easily modified and assembled exploiting proper connecting linkers of different nature. Thus, substantial research efforts have been put in the construction of dimeric/multimeric versions of effective aptamers with various degrees of success in target binding affinity or therapeutic activity enhancement. The present review summarizes recent advances in the design and development of dimeric and multimeric DNA-based aptamers, including those forming G-quadruplex (G4) structures, recognizing different key proteins in relevant pathological processes. Most of the designed constructs have shown improved performance in terms of binding affinity or therapeutic activity as anti-inflammatory, antiviral, anticoagulant, and anticancer agents and their number is certainly bound to grow in the next future.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, via Sergio Pansini, 5, I-80131 Naples, Italy
| | - Ettore Napolitano
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
- Institute of Biostructures and Bioimages, CNR, via Mezzocannone 16, I-80134 Naples, Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; (E.N.); (D.M.); (D.M.)
| |
Collapse
|
17
|
Riccardi C, Napolitano E, Platella C, Musumeci D, Melone MAB, Montesarchio D. Anti-VEGF DNA-based aptamers in cancer therapeutics and diagnostics. Med Res Rev 2020; 41:464-506. [PMID: 33038031 DOI: 10.1002/med.21737] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/12/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
The vascular endothelial growth factor (VEGF) family and its receptors play fundamental roles not only in physiological but also in pathological angiogenesis, characteristic of cancer progression. Aiming at finding putative treatments for several malignancies, various small molecules, antibodies, or protein-based drugs have been evaluated in vitro and in vivo as VEGF inhibitors, providing efficient agents approved for clinical use. Due to the high clinical importance of VEGF, also a great number of anti-VEGF nucleic acid-based aptamers-that is, oligonucleotides able to bind with high affinity and specificity a selected biological target-have been developed as promising agents in anticancer strategies. Notable research efforts have been made in optimization processes of the identified aptamers, searching for increased target affinity and/or bioactivity by exploring structural analogues of the lead compounds. This review is focused on recent studies devoted to the development of DNA-based aptamers designed to target VEGF. Their therapeutic potential as well as their significance in the construction of highly selective biosensors is here discussed.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.,Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Inter-University Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ettore Napolitano
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.,Institute of Biostructures and Bioimages, Naples, Italy
| | - Mariarosa A B Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Inter-University Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
18
|
Riccardi C, Napolitano E, Platella C, Musumeci D, Montesarchio D. G-quadruplex-based aptamers targeting human thrombin: Discovery, chemical modifications and antithrombotic effects. Pharmacol Ther 2020; 217:107649. [PMID: 32777331 DOI: 10.1016/j.pharmthera.2020.107649] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
First studies on thrombin-inhibiting DNA aptamers were reported in 1992, and since then a large number of anticoagulant aptamers has been discovered. TBA - also named HD1, a 15-mer G-quadruplex (G4)-forming oligonucleotide - is the best characterized thrombin binding aptamer, able to specifically recognize the protein exosite I, thus inhibiting the conversion of soluble fibrinogen into insoluble fibrin strands. Unmodified nucleic acid-based aptamers, in general, and TBA in particular, exhibit limited pharmacokinetic properties and are rapidly degraded in vivo by nucleases. In order to improve the biological performance of aptamers, a widely investigated strategy is the introduction of chemical modifications in their backbone at the level of the nucleobases, sugar moieties or phosphodiester linkages. Besides TBA, also other thrombin binding aptamers, able to adopt a well-defined G4 structure, e.g. mixed duplex/quadruplex sequences, as well as homo- and hetero-bivalent constructs, have been identified and optimized. Considering the growing need of new efficient anticoagulant agents associated with the strong therapeutic potential of these thrombin inhibitors, the research on thrombin binding aptamers is still a very hot and intriguing field. Herein, we comprehensively described the state-of-the-art knowledge on the DNA-based aptamers targeting thrombin, especially focusing on the optimized analogues obtained by chemically modifying the oligonucleotide backbone, and their biological performances in therapeutic applications.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; Department of Advanced Medical and Surgical Sciences, 2(nd) Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, via Sergio Pansini, 5, I-80131 Naples, Italy.
| | - Ettore Napolitano
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy.
| | - Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy.
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy; Institute of Biostructures and Bioimages, CNR, via Mezzocannone 16, I-80134 Naples, Italy.
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21, I-80126 Naples, Italy.
| |
Collapse
|
19
|
Manochehry S, Gu J, McConnell EM, Salena BJ, Li Y. In Vitro Selection of New DNA Aptamers for Human Vascular Endothelial Growth Factor 165. Chembiochem 2020; 21:2029-2036. [PMID: 32180322 DOI: 10.1002/cbic.202000024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/26/2020] [Indexed: 11/06/2022]
Abstract
Two DNA aptamers that bind the heparin-binding domain (HBD) of the human vascular endothelial growth factor 165 (VEGF-165) have been previously reported. Although VEGF-165 is a homodimeric protein and the two aptamers have different sequences and secondary structures, the aptamers appear to occupy the same binding site and cannot form a 2 : 1 aptamer/protein complex, thus making them unsuitable for creating a higher-affinity dimeric DNA aptamer. This has motivated us to conduct a new in vitro selection experiment to search for new VEGF-165-binding DNA aptamers with different properties. We undertook a multistream selection strategy in which the concentration of VEGF-165 was varied significantly. We carried out 11 rounds of selection, and next-generation sequencing was conducted for every round in each stream. From comprehensive sequence analysis, we identified four classes of DNA aptamers, of which two were reported before, but two are new DNA aptamers. One of the new aptamers exhibits a unique property that has never been observed before: it is capable of forming the 2 : 1 aptamer/protein complex with VEGF-165. This work has expanded the repertoire of VEGF-165-binding DNA aptamers and creates a possibility to engineer a higher affinity homodimeric aptamer for VEGF-165.
Collapse
Affiliation(s)
- Sepehr Manochehry
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Jimmy Gu
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Erin M McConnell
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Bruno J Salena
- Department of Medicine DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| | - Yingfu Li
- M.G. DeGroote Institute for Infectious Disease Research Department of Biochemistry and Biomedical Sciences DeGroote School of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4 K1, Canada
| |
Collapse
|
20
|
Riccardi C, Musumeci D, Platella C, Gaglione R, Arciello A, Montesarchio D. Tuning the Polymorphism of the Anti-VEGF G-rich V7t1 Aptamer by Covalent Dimeric Constructs. Int J Mol Sci 2020; 21:ijms21061963. [PMID: 32183039 PMCID: PMC7139925 DOI: 10.3390/ijms21061963] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 01/11/2023] Open
Abstract
In the optimization process of nucleic acid aptamers, increased affinity and/or activity are generally searched by exploring structural analogues of the lead compound. In many cases, promising results have been obtained by dimerization of the starting aptamer. Here we studied a focused set of covalent dimers of the G-quadruplex (G4) forming anti-Vascular Endothelial Growth Factor (VEGF) V7t1 aptamer with the aim of identifying derivatives with improved properties. In the design of these covalent dimers, connecting linkers of different chemical nature, maintaining the same polarity along the strand or inverting it, have been introduced. These dimeric aptamers have been investigated using several biophysical techniques to disclose the conformational behavior, molecularity and thermal stability of the structures formed in different buffers. This in-depth biophysical characterization revealed the formation of stable G4 structures, however in some cases accompanied by alternative tridimensional arrangements. When tested for their VEGF165 binding and antiproliferative activity in comparison with V7t1, these covalent dimers showed slightly lower binding ability to the target protein but similar if not slightly higher antiproliferative activity on human breast adenocarcinoma MCF-7 cells. These results provide useful information for the design of improved dimeric aptamers based on further optimization of the linker joining the two consecutive V7t1 sequences.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; (C.R.); (D.M.); (C.P.); (R.G.); (A.A.)
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; (C.R.); (D.M.); (C.P.); (R.G.); (A.A.)
- Institute of Biostructures and Bioimages (IBB), CNR, Via Mezzocannone 16, I-80134 Napoli, Italy
| | - Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; (C.R.); (D.M.); (C.P.); (R.G.); (A.A.)
| | - Rosa Gaglione
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; (C.R.); (D.M.); (C.P.); (R.G.); (A.A.)
| | - Angela Arciello
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; (C.R.); (D.M.); (C.P.); (R.G.); (A.A.)
- National Institute of Biostructures and Biosystems (INBB), 00136 Rome, Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; (C.R.); (D.M.); (C.P.); (R.G.); (A.A.)
- Correspondence:
| |
Collapse
|