151
|
Polyclonal Aptamers for Specific Fluorescence Labeling and Quantification of the Health Relevant Human Gut Bacterium Parabacteroides distasonis. Microorganisms 2021; 9:microorganisms9112284. [PMID: 34835410 PMCID: PMC8618460 DOI: 10.3390/microorganisms9112284] [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: 09/28/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/27/2022] Open
Abstract
Single-stranded DNA aptamers as affinity molecules for the rapid, reliable detection of intestinal bacteria are of particular interest to equip health systems with novel robust and cheap diagnostic tools for monitoring the success of supplementation strategies with selected probiotic gut bacteria in the fight against major widespread threats, such as obesity and neurodegenerative diseases. The human gut bacterium Parabacteroides distasonis (P. distasonis) is positively associated with diseases such as obesity, non-alcoholic fatty liver disease and multiple sclerosis with reduced cell counts in these diseases and is thus a promising potential probiotic bacterium for future microbial supplementation. In this paper we report on the evolution of a specific polyclonal aptamer library by the fluorescence based FluCell-SELEX directed against whole cells of P. distasonis that specifically and efficiently binds and labels P. distasonis. The aptamer library showed high binding affinity and was suited to quantitatively discriminate P. distasonis from other prominent gut bacteria also in mixtures. We believe that this library against a promising probiotic bacterium as a prototype may open new routes towards the development of novel biosensors for the easy and efficient quantitative monitoring of microbial abundance in human microbiomes in general.
Collapse
|
152
|
Cen Y, Wang Z, Ke P, Zhu W, Yuan Z, Feng S, Chen Y, Lin C, Liu X, Li Y, Yan P. Development of a novel ssDNA aptamer targeting cardiac troponin I and its clinical applications. Anal Bioanal Chem 2021; 413:7043-7053. [PMID: 34673993 DOI: 10.1007/s00216-021-03667-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/30/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
Cardiac troponin I (cTnI) is a specific biomarker of acute myocardial infarction (AMI). However, cTnI detection kits prepared with antibodies have many defects. Nucleic acid aptamers are sequences of single-strand DNA or RNA that can overcome the deficiency of antibodies. Herein, sandwich ELONA methods were established based on aptamers. Two selected ssDNA aptamers (Apt3 and Apt6) showed high binding affinity and sensibility (Apt3: Kd = 1.01 ± 0.07 nM, Apt6: k = 0.68 ± 0.05) and did not bind to the same domain of cTnI. Therefore, these two aptamers can be applied to the ELONA methods. The detection range of cTnI using the dual-aptamer sandwich ELONA method was 0.05-200 ng/mL, and the bioanalytical method verification results can meet the national standard of Chinese Pharmacopoeia (2020 Edition). There was no difference between results of the dual-aptamer sandwich ELONA method and the diagnostic results of serum obtained from 243 people (P = 0.39, P ˃ 0.05). The sensitivity and specificity of the ELONA with cTnI in serum were 96.46% and 93.85%, respectively. Compared with the FICA kit, which is clinically used, the consequences of ELONA method are closer to the diagnostic results. This study suggests that the aptamers Apt3 and Apt6 have high affinity and strong specificity and that the dual-aptamer sandwich ELONA method has a wide detection range and can be used to determine cTnI in serum, with potential applications in the diagnosis of AMIs.
Collapse
Affiliation(s)
- Yi Cen
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhongping Wang
- Tongpeng Zhongxu Pharmaceutical Technology Company, Guangzhou, Guangdong, China
| | - Peixiong Ke
- Department of Pharmacology, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenting Zhu
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhongwen Yuan
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Senling Feng
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiqing Chen
- Tongpeng Zhongxu Pharmaceutical Technology Company, Guangzhou, Guangdong, China
| | - Caiyan Lin
- Tongpeng Zhongxu Pharmaceutical Technology Company, Guangzhou, Guangdong, China
| | - Xiaomin Liu
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuting Li
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pengke Yan
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
153
|
Yuan R, Li HK, He H. Recent advances in metal/covalent organic framework-based electrochemical aptasensors for biosensing applications. Dalton Trans 2021; 50:14091-14104. [PMID: 34609402 DOI: 10.1039/d1dt02360h] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The booming development of novel porous materials, metal-organic frameworks (MOFs) and covalent-organic frameworks (COFs) has been attracting a lot of attention due to their designabilities, diversities, and extensive applications. MOFs and COFs provide a new potential opportunity and platform to fabricate electrochemical aptasensors for biosensing applications. Compared to other traditional materials, MOF/COF-based electrochemical biosensors can appreciably amplify the electrochemical response signals to improve the sensing performance. Herein, we provide a comprehensive overview of MOF/COF-based electrochemical aptasensors for monitoring different ultra-trace analytes (e.g. antibiotics, pesticides, and cancer markers). This review systematically discusses the classification of electrochemical aptasensors based on various functional materials, including pure MOFs, MOF/conductive composites, metal nanoparticle/MOF composites, pure COFs, COFs/conductive composites, and other hybrid materials. Furthermore, some typical MOF/COF-based electrochemical aptasensors in the recognition of specific targets are described in detail to improve and guide further research for biosensing applications.
Collapse
Affiliation(s)
- Rongrong Yuan
- Department of Materials Science and Engineering, Jilin Jianzhu University, Changchun 130118, P. R. China
| | - Hong-Kai Li
- Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China.
| | - Hongming He
- Tianjin Key Laboratory of Structure and Performance for Functional Molecules, College of Chemistry, Tianjin Normal University, Tianjin 300387, P. R. China.
| |
Collapse
|
154
|
Li R, Wen Y, Wang F, He P. Recent advances in immunoassays and biosensors for mycotoxins detection in feedstuffs and foods. J Anim Sci Biotechnol 2021; 12:108. [PMID: 34629116 PMCID: PMC8504128 DOI: 10.1186/s40104-021-00629-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/08/2021] [Indexed: 12/22/2022] Open
Abstract
Mycotoxins are secondary metabolites produced by fungus. Many mycotoxin species are highly toxic and are frequently found in cereals and feedstuffs. So, powerful detection methods are vital and effective ways to prevent feed contamination. Traditional detection methods can no longer meet the needs of massive, real-time, simple, and fast mycotoxin monitoring. Rapid detection methods based on advanced material and sensor technology are the future trend. In this review, we highlight recent progress of mycotoxin rapid detection strategies in feedstuffs and foods, especially for simultaneous multiplex mycotoxin determination. Immunoassays, biosensors, and the prominent roles of nanomaterials are introduced. The principles of different types of recognition and signal transduction are explained, and the merits and pitfalls of these methods are compared. Furthermore, limitations and challenges of existing rapid sensing strategies and perspectives of future research are discussed.
Collapse
Affiliation(s)
- Runxian Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yang Wen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Fenglai Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Pingli He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
155
|
Burtscher B, Manco Urbina PA, Diacci C, Borghi S, Pinti M, Cossarizza A, Salvarani C, Berggren M, Biscarini F, Simon DT, Bortolotti CA. Sensing Inflammation Biomarkers with Electrolyte-Gated Organic Electronic Transistors. Adv Healthc Mater 2021; 10:e2100955. [PMID: 34423579 PMCID: PMC11469060 DOI: 10.1002/adhm.202100955] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/16/2021] [Indexed: 01/08/2023]
Abstract
An overview of cytokine biosensing is provided, with a focus on the opportunities provided by organic electronic platforms for monitoring these inflammation biomarkers which manifest at ultralow concentration levels in physiopathological conditions. Specifically, two of the field's state-of-the-art technologies-organic electrochemical transistors (OECTs) and electrolyte gated organic field effect transistors (EGOFETs)-and their use in sensing cytokines and other proteins associated with inflammation are a particular focus. The overview will include an introduction to current clinical and "gold standard" quantification techniques and their limitations in terms of cost, time, and required infrastructure. A critical review of recent progress with OECT- and EGOFET-based protein biosensors is presented, alongside a discussion onthe future of these technologies in the years and decades ahead. This is especially timely as the world grapples with limited healthcare diagnostics during the Coronavirus disease (COVID-19)pandemic where one of the worst-case scenarios for patients is the "cytokine storm." Clearly, low-cost point-of-care technologies provided by OECTs and EGOFETs can ease the global burden on healthcare systems and support professionals by providing unprecedented wealth of data that can help to monitor disease progression in real time.
Collapse
Affiliation(s)
- Bernhard Burtscher
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | | | - Chiara Diacci
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Simone Borghi
- Department of Life SciencesUniversity of Modena and Reggio EmiliaVia Campi 103Modena41125Italy
| | - Marcello Pinti
- Department of Life SciencesUniversity of Modena and Reggio EmiliaVia Campi 103Modena41125Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and AdultsUniversity of Modena and Reggio EmiliaVia Campi 287Modena41125Italy
| | - Carlo Salvarani
- Rheumatology UnitUniversity of Modena and Reggio EmiliaMedical SchoolAzienda Ospedaliero‐UniversitariaPoliclinico di ModenaModena41124Italy
| | - Magnus Berggren
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Fabio Biscarini
- Department of Life SciencesUniversity of Modena and Reggio EmiliaVia Campi 103Modena41125Italy
- Center for Translation NeurophysiologyIstituto Italiano di TecnologiaVia Fossato di Mortara 17–19Ferrara44100Italy
| | - Daniel T. Simon
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Carlo A. Bortolotti
- Department of Life SciencesUniversity of Modena and Reggio EmiliaVia Campi 103Modena41125Italy
| |
Collapse
|
156
|
Zhong J, Ding J, Deng L, Xiang Y, Liu D, Zhang Y, Chen X, Yang Q. Selection of DNA Aptamers Recognizing EpCAM-Positive Prostate Cancer by Cell-SELEX for in vitro and in vivo MR Imaging. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3985-3996. [PMID: 34584404 PMCID: PMC8464308 DOI: 10.2147/dddt.s322854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/06/2021] [Indexed: 11/23/2022]
Abstract
Background The sensitive and specific detection of pathogenic cells is important in tumor diagnosis at an early stage. Aptamers are short single-stranded oligonucleotides evolved from systematic evolution of ligands by exponential enrichment (SELEX). It has been proved that aptamers can interact with cognate target molecules with high affinity and specificity and have great potential in the development of medical imaging at molecular level. Purpose To select epithelial cell adhesion molecule (EpCAM) specific aptamers targeting prostate cancer and further to conjugate aptamers with GoldMag nanoparticles (a typical iron oxide core/gold shell structure) to construct magnetic molecular probes for medical imaging. Methods EpCAM-specific aptamers were selected by Cell-SELEX. The enrichment of specific aptamer candidates was monitored by flow cytometric analysis. Aptamers were further conjugated with GoldMag nanoparticles to construct magnetic molecular probes. The affinity and specificity of aptamer candidates and aptamer-conjugated GoldMag nanoparticles were evaluated. The MR imaging of aptamer-conjugated GoldMag nanoparticles to prostate cancer was further explored in vitro and in vivo. Results After 12 rounds of selection, aptamer candidates Eppc6 and Eppc14 could specifically target three types of prostate cancer cells, revealing a high affinity of Eppc6 and Eppc14. Moreover, aptamer-conjugated GoldMag nanoparticles not only exhibited good affinity to different prostate cancer cells but also produced strong T2WI signal intensity reduction distinguished from peritumoral tissue in MRI, indicating that the molecular probes possess both the affinity properties of EpCAM-specific aptamer and the superparamagnetic features of iron oxide. Conclusion Our study indicates that aptamer Eppc6 and Eppc14 can recognize prostate cancer cells and tissues. The aptamer-conjugated GoldMag nanoparticles constructed in the study can be used as a molecular imaging agent for detection of PCa in MRI.
Collapse
Affiliation(s)
- Jinman Zhong
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Jianke Ding
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, People's Republic of China
| | - Lei Deng
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Ying Xiang
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Duoduo Liu
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Yanyan Zhang
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Xin Chen
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| | - Quanxin Yang
- Department of Radiology, The Second Affiliated Hospital, Xi' an Jiaotong University, Xi'an, Shaanxi Province, 710004, People's Republic of China
| |
Collapse
|
157
|
Khan S, Burciu B, Filipe CDM, Li Y, Dellinger K, Didar TF. DNAzyme-Based Biosensors: Immobilization Strategies, Applications, and Future Prospective. ACS NANO 2021; 15:13943-13969. [PMID: 34524790 DOI: 10.1021/acsnano.1c04327] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Since their discovery almost three decades ago, DNAzymes have been used extensively in biosensing. Depending on the type of DNAzyme being used, these functional oligonucleotides can act as molecular recognition elements within biosensors, offering high specificity to their target analyte, or as reporters capable of transducing a detectable signal. Several parameters need to be considered when designing a DNAzyme-based biosensor. In particular, given that many of these biosensors immobilize DNAzymes onto a sensing surface, selecting an appropriate immobilization strategy is vital. Suboptimal immobilization can result in both DNAzyme detachment and poor accessibility toward the target, leading to low sensing accuracy and sensitivity. Various approaches have been employed for DNAzyme immobilization within biosensors, ranging from amine and thiol-based covalent attachment to non-covalent strategies involving biotin-streptavidin interactions, DNA hybridization, electrostatic interactions, and physical entrapment. While the properties of each strategy inform its applicability within a proposed sensor, the selection of an appropriate strategy is largely dependent on the desired application. This is especially true given the diverse use of DNAzyme-based biosensors for the detection of pathogens, metal ions, and clinical biomarkers. In an effort to make the development of such sensors easier to navigate, this paper provides a comprehensive review of existing immobilization strategies, with a focus on their respective advantages, drawbacks, and optimal conditions for use. Next, common applications of existing DNAzyme-based biosensors are discussed. Last, emerging and future trends in the development of DNAzyme-based biosensors are discussed, and gaps in existing research worthy of exploration are identified.
Collapse
Affiliation(s)
- Shadman Khan
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Brenda Burciu
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, North Carolina 27401, United States
| | - Carlos D M Filipe
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, North Carolina 27401, United States
| | - Tohid F Didar
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L7, Canada
| |
Collapse
|
158
|
Beyond G-Quadruplexes-The Effect of Junction with Additional Structural Motifs on Aptamers Properties. Int J Mol Sci 2021; 22:ijms22189948. [PMID: 34576112 PMCID: PMC8466185 DOI: 10.3390/ijms22189948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/02/2022] Open
Abstract
G-quadruplexes constitute an important type of nucleic acid structure, which can be found in living cells and applied by cell machinery as pivotal regulatory elements. Importantly, robust development of SELEX technology and modern, nucleic acid-based therapeutic strategies targeted towards various molecules have also revealed a large group of potent aptamers whose structures are grounded in G-quadruplexes. In this review, we analyze further extension of tetraplexes by additional structural elements and investigate whether G-quadruplex junctions with duplex, hairpin, triplex, or second G-quadruplex motifs are favorable for aptamers stability and biological activity. Furthermore, we indicate the specific and pivotal role of the G-quadruplex domain and the additional structural elements in interactions with target molecules. Finally, we consider the potency of G-quadruplex junctions in future applications and indicate the emerging research area that is still waiting for development to obtain highly specific and effective nucleic acid-based molecular tools.
Collapse
|
159
|
Cleri F, Lensink MF, Blossey R. DNA Aptamers Block the Receptor Binding Domain at the Spike Protein of SARS-CoV-2. Front Mol Biosci 2021; 8:713003. [PMID: 34458322 PMCID: PMC8397481 DOI: 10.3389/fmolb.2021.713003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/28/2021] [Indexed: 12/22/2022] Open
Abstract
DNA aptamers are versatile molecular species obtained by the folding of short single-stranded nucleotide sequences, with highly specific recognition capabilities against proteins. Here we test the ability of DNA aptamers to interact with the spike (S-)protein of the SARS-CoV-2 viral capsid. The S-protein, a trimer made up of several subdomains, develops the crucial function of recognizing the ACE2 receptors on the surface of human cells, and subsequent fusioning of the virus membrane with the host cell membrane. In order to achieve this, the S1 domain of one protomer switches between a closed conformation, in which the binding site is inaccessible to the cell receptors, and an open conformation, in which ACE2 can bind, thereby initiating the entry process of the viral genetic material in the host cell. Here we show, by means of state-of-the-art molecular simulations, that small DNA aptamers experimentally identified can recognize the S-protein of SARS-CoV-2, and characterize the details of the binding process. We find that their interaction with different subdomains of the S-protein can effectively block, or at least considerably slow down the opening process of the S1 domain, thereby significantly reducing the probability of virus-cell binding. We provide evidence that, as a consequence, binding of the human ACE2 receptor may be crucially affected under such conditions. Given the facility and low cost of fabrication of specific aptamers, the present findings could open the way to both an innovative viral screening technique with sub-nanomolar sensitivity, and to an effective and low impact curative strategy.
Collapse
Affiliation(s)
- Fabrizio Cleri
- University of Lille, CNRS UMR8520 IEMN, Institut d’Electronique, Microélectronique et Nanotechnologie, Lille, France
- University of Lille, Departement de Physique, Villeneuve d’Ascq, France
| | - Marc F. Lensink
- University of Lille, CNRS UMR8576 UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Ralf Blossey
- University of Lille, CNRS UMR8576 UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| |
Collapse
|
160
|
Zou X, Wu J, Gu J, Shen L, Mao L. DNA aptamer against EV-A71 VP1 protein: selection and application. Virol J 2021; 18:164. [PMID: 34384436 PMCID: PMC8359077 DOI: 10.1186/s12985-021-01631-y] [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: 06/01/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Enterovirus 71 (EV-A71) is a highly infectious pathogen associated with hand, foot and mouth disease, herpangina, and various neurological complications, so it is important for the early detection and treatment of EV-A71. An aptamer is a nucleotide sequence that screened in vitro by the technology named systematic evolution of ligands by exponential enrichment technology (SELEX). Similar to antibodies, aptamers can bind to the targets with high specificity and affinity. Besides, emerging aptamers have many advantages comparing with antibodies, such as ease of synthesis and modification, having a wide variety of target materials, low manufacturing cost and easy flexibility in amending. Therefore, aptamers are promising in virus detection and anti-virus therapy. METHODS Aptamers were selected by SELEX. Specificity, affinity and second structure were used to characterize the selected aptamers. Chemiluminescence was adopted to build an aptamer-based detection method for EV-A71. Cytopathogenic effects trial, the level of intracellular EV-A71 RNA and protein expression were used to evaluate the antiviral effect of the selected aptamers. RESULTS Three DNA aptamers with high specificity and affinity for EV-A71structual protein VP1 were screened out. A rapid chemiluminutesescence aptamer biosensor for EV-A71 detection was designed out. The selected aptamers could inhibit the RNA replication and protein expression of EV-A71 in RD cells and ameliorate the cytopathogenic effects. CONCLUSIONS The aptamers against EV-A71 have the potentiality to be applied as attractive candidates used for EV-A71 detection and treatment in the future.
Collapse
Affiliation(s)
- Xinran Zou
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang City, Jiangsu Province, China.,Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou City, China
| | - Jing Wu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Jiaqi Gu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Li Shen
- Department of Laboratory, Zhenjiang Center for Disease Control and Prevention, Zhenjiang City, Jiangsu Province, China
| | - Lingxiang Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang City, Jiangsu Province, China.
| |
Collapse
|
161
|
Jiang L, Qu X, Sun W, Zhang M, Wang Y, Wang Y, Zhao Y, Zhang F, Leng Y, Liu S, Yu J, Huang J. A three-dimensional dynamic DNA walker-mediated branching hybridization chain reaction for the ultrasensitive fluorescence sensing of ampicillin. Analyst 2021; 146:5413-5420. [PMID: 34346408 DOI: 10.1039/d0an02226h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In this study, a novel, rapid and ultrasensitive fluorescence strategy using the three-dimensional (3D) dynamic DNA walker (DW)-induced branched hybridization chain reaction (bHCR) has been proposed for the detection of ampicillin (AMP). The sensing system was composed of an Nt·Bbvcl-powered DNA walker blocked by an AMP aptamer, hairpin-shaped DNA track probe (TP) and four kinds of metastable hairpin probes as the substrates of bHCR, which triggered the formation of the split G-quadruplex as the signal molecule. Due to the reasonable design, the specific binding between AMP and its aptamer activated the DW, and the DW moved on the surface of the gold nanoparticles (AuNPs) with the help of Nt·Bbvcl to produce primer probes (PPs), which induced bHCR. The products of the bHCR gathered two split G-quadruplex sequences together to form one complete G-quadruplex. The formed G-quadruplex emitted a strong fluorescence signal in the presence of thioflavin-T (ThT) to achieve the purpose of detecting AMP. The sensitivity of this method was greatly improved by the use of the 3D DNA walker and bHCR. The split G-quadruplex enhanced the signal-to-noise ratio (SNR). Under the optimal experimental conditions, a good correlation was obtained between the fluorescence intensity of the sensing system and the concentration of AMP ranging from 5 pM to 500 nM with a limit of detection (LOD) of 3.68 pM. Simultaneously, the method has been applied to the detection of antibiotics in spiked milk samples with satisfactory results.
Collapse
Affiliation(s)
- Long Jiang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Jing L, Qin M, Zhang X, Song Y, Zhang J, Xia X, Gao K, Han Q. A novel borax-specific ssDNA aptamer screened by high-throughput SELEX and its colorimetric assay with aggregation of AuNPs. J Food Compost Anal 2021. [DOI: 10.1016/j.jfca.2021.103947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
163
|
Martin DR, Sibuyi NR, Dube P, Fadaka AO, Cloete R, Onani M, Madiehe AM, Meyer M. Aptamer-Based Diagnostic Systems for the Rapid Screening of TB at the Point-of-Care. Diagnostics (Basel) 2021; 11:1352. [PMID: 34441287 PMCID: PMC8391981 DOI: 10.3390/diagnostics11081352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022] Open
Abstract
The transmission of Tuberculosis (TB) is very rapid and the burden it places on health care systems is felt globally. The effective management and prevention of this disease requires that it is detected early. Current TB diagnostic approaches, such as the culture, sputum smear, skin tuberculin, and molecular tests are time-consuming, and some are unaffordable for low-income countries. Rapid tests for disease biomarker detection are mostly based on immunological assays that use antibodies which are costly to produce, have low sensitivity and stability. Aptamers can replace antibodies in these diagnostic tests for the development of new rapid tests that are more cost effective; more stable at high temperatures and therefore have a better shelf life; do not have batch-to-batch variations, and thus more consistently bind to a specific target with similar or higher specificity and selectivity and are therefore more reliable. Advancements in TB research, in particular the application of proteomics to identify TB specific biomarkers, led to the identification of a number of biomarker proteins, that can be used to develop aptamer-based diagnostic assays able to screen individuals at the point-of-care (POC) more efficiently in resource-limited settings.
Collapse
Affiliation(s)
- Darius Riziki Martin
- DSI/Mintek Nanotechnology Innovation Centre-Biolabels Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (D.R.M.); (N.R.S.); (P.D.); (A.O.F.); (A.M.M.)
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa;
| | - Nicole Remaliah Sibuyi
- DSI/Mintek Nanotechnology Innovation Centre-Biolabels Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (D.R.M.); (N.R.S.); (P.D.); (A.O.F.); (A.M.M.)
| | - Phumuzile Dube
- DSI/Mintek Nanotechnology Innovation Centre-Biolabels Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (D.R.M.); (N.R.S.); (P.D.); (A.O.F.); (A.M.M.)
| | - Adewale Oluwaseun Fadaka
- DSI/Mintek Nanotechnology Innovation Centre-Biolabels Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (D.R.M.); (N.R.S.); (P.D.); (A.O.F.); (A.M.M.)
| | - Ruben Cloete
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa;
| | - Martin Onani
- Department of Chemistry, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa;
| | - Abram Madimabe Madiehe
- DSI/Mintek Nanotechnology Innovation Centre-Biolabels Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (D.R.M.); (N.R.S.); (P.D.); (A.O.F.); (A.M.M.)
| | - Mervin Meyer
- DSI/Mintek Nanotechnology Innovation Centre-Biolabels Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville 7535, South Africa; (D.R.M.); (N.R.S.); (P.D.); (A.O.F.); (A.M.M.)
| |
Collapse
|
164
|
Ouyang Y, Biniuri Y, Fadeev M, Zhang P, Carmieli R, Vázquez-González M, Willner I. Aptamer-Modified Cu 2+-Functionalized C-Dots: Versatile Means to Improve Nanozyme Activities-"Aptananozymes". J Am Chem Soc 2021; 143:11510-11519. [PMID: 34286967 PMCID: PMC8856595 DOI: 10.1021/jacs.1c03939] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
The covalent linkage of aptamer binding
sites to nanoparticle nanozymes
is introduced as a versatile method to improve the catalytic activity
of nanozymes by concentrating the reaction substrates at the catalytic
nanozyme core, thereby emulating the binding and catalytic active-site
functions of native enzymes. The concept is exemplified with the synthesis
of Cu2+ ion-functionalized carbon dots (C-dots), modified
with the dopamine binding aptamer (DBA) or the tyrosinamide binding
aptamer (TBA), for the catalyzed oxidation of dopamine to aminochrome
by H2O2 or the oxygenation of l-tyrosinamide
to the catechol product, which is subsequently oxidized to amidodopachrome,
in the presence of H2O2/ascorbate mixture. Sets
of structurally functionalized DBA-modified Cu2+ ion-functionalized
C-dots or sets of structurally functionalized TBA-modified Cu2+ ion-functionalized C-dots are introduced as nanozymes of
superior catalytic activities (aptananozymes) toward the oxidation
of dopamine or the oxygenation of l-tyrosinamide, respectively.
The aptananozymes reveal enhanced catalytic activities as compared
to the separated catalyst and respective aptamer constituents. The
catalytic functions of the aptananozymes are controlled by the structure
of the aptamer units linked to the Cu2+ ion-functionalized
C-dots. In addition, the aptananozyme shows chiroselective catalytic
functions demonstrated by the chiroselective-catalyzed oxidation of l/d-DOPA to l/d-dopachrome. Binding
studies of the substrates to the different aptananozymes and mechanistic
studies associated with the catalytic transformations are discussed.
Collapse
Affiliation(s)
- Yu Ouyang
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Yonatan Biniuri
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Michael Fadeev
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Pu Zhang
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Raanan Carmieli
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Itamar Willner
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
165
|
Wang Y, Chen L, Xuan T, Wang J, Wang X. Label-free Electrochemical Impedance Spectroscopy Aptasensor for Ultrasensitive Detection of Lung Cancer Biomarker Carcinoembryonic Antigen. Front Chem 2021; 9:721008. [PMID: 34350159 PMCID: PMC8326396 DOI: 10.3389/fchem.2021.721008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/07/2021] [Indexed: 01/13/2023] Open
Abstract
In this work, an integrated electrode system consisting of a graphene working electrode, a carbon counter electrode and an Ag/AgCl reference electrode was fabricated on an FR-4 glass fiber plate by a polyethylene self-adhesive mask stencil method combined with a manual screen printing technique. The integrated graphene electrode was used as the base electrode, and AuNPs were deposited on the working electrode surface by cyclic voltammetry. Then, the carcinoembryonic antigen aptamer was immobilized using the sulfhydryl self-assembly technique. The sensor uses [Fe(CN)6]3-/4- as a redox probe for label free detection of carcinoembryonic antigen based on the impedance change caused by the difference in electron transfer rate before and after the binding of carcinoembryonic antigen aptamer and the target carcinoembryonic antigen. The results showed a good linear relationship when the CEA concentration is in the range of 0.2-15.0 ng/ml. The detection limit was calculated to be 0.085 ng/ml (S/N = 3).
Collapse
Affiliation(s)
- Yawei Wang
- Department of Medical Oncology, Qilu Hospital, Shandong University, Jinan, China
| | - Lei Chen
- Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Tiantian Xuan
- Department of Medical Oncology, Qilu Hospital, Shandong University, Jinan, China
| | - Jian Wang
- Department of Medical Oncology, Qilu Hospital, Shandong University, Jinan, China
| | - Xiuwen Wang
- Department of Medical Oncology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
166
|
Liu L, Han Z, An F, Gong X, Zhao C, Zheng W, Mei L, Zhou Q. Aptamer-based biosensors for the diagnosis of sepsis. J Nanobiotechnology 2021; 19:216. [PMID: 34281552 PMCID: PMC8287673 DOI: 10.1186/s12951-021-00959-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022] Open
Abstract
Sepsis, the syndrome of infection complicated by acute organ dysfunction, is a serious and growing global problem, which not only leads to enormous economic losses but also becomes one of the leading causes of mortality in the intensive care unit. The detection of sepsis-related pathogens and biomarkers in the early stage plays a critical role in selecting appropriate antibiotics or other drugs, thereby preventing the emergence of dangerous phases and saving human lives. There are numerous demerits in conventional detection strategies, such as high cost, low efficiency, as well as lacking of sensitivity and selectivity. Recently, the aptamer-based biosensor is an emerging strategy for reasonable sepsis diagnosis because of its accessibility, rapidity, and stability. In this review, we first introduce the screening of suitable aptamer. Further, recent advances of aptamer-based biosensors in the detection of bacteria and biomarkers for the diagnosis of sepsis are summarized. Finally, the review proposes a brief forecast of challenges and future directions with highly promising aptamer-based biosensors.
Collapse
Affiliation(s)
- Lubin Liu
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Zeyu Han
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Fei An
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Xuening Gong
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Chenguang Zhao
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Weiping Zheng
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Li Mei
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Qihui Zhou
- Institute for Translational Medicine, Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China.
- School of Stomatology, Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
167
|
Song J, Lin X, Jiang N, Huang M. Carbon-doped WO 3 electrochemical aptasensor based on Box-Behnken strategy for highly-sensitive detection of tetracycline. Food Chem 2021; 367:130564. [PMID: 34365249 DOI: 10.1016/j.foodchem.2021.130564] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 02/05/2023]
Abstract
Aptamer has been proved to be an important probe for antibiotic detection. Here, the electrical signal was doubly amplified by the synergistic effect of C-WO3 and AuNPs. The probe structure has a specific recognition effect on tetracycline, which improves the selectivity and anti-interference of the sensor. With the assistance of BBD strategy, the experimental errors of the C-WO3@AuNPs aptasensor were reduced and the best conditions for its preparation were obtained. This was conducive to obtain the best electrical signal transmission capacity of the electrode, greatly improved the sensor sensitivity. Under this mechanism, the antibiotic sensor achieved a low detection range (0.1 nM-100 nM) and a low detection limit (4.8 × 10-2 nM). The sensor showed excellent selectivity even in the presence of coexisting pollutants. This work explored the mechanism of charge change and demonstrated the role of probes in antibiotic sensing, providing important prospects of future applications in electrochemical sensors.
Collapse
Affiliation(s)
- Jialing Song
- College of Environmental Science and Engineering, State Environmental Protection Engineering Center for Pollution Treatment and Control in Textile Industry, Donghua University, Shanghai 201620, China; National University of Singapore, Department of Chemistry, 3 Science Drive 3, Singapore 117543, Singapore
| | - Xuanhao Lin
- National University of Singapore, Department of Chemistry, 3 Science Drive 3, Singapore 117543, Singapore
| | - Nan Jiang
- College of Environmental Science and Engineering, State Environmental Protection Engineering Center for Pollution Treatment and Control in Textile Industry, Donghua University, Shanghai 201620, China
| | - Manhong Huang
- College of Environmental Science and Engineering, State Environmental Protection Engineering Center for Pollution Treatment and Control in Textile Industry, Donghua University, Shanghai 201620, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China; Donghua Uni, Key Lab Sci & Technol Ecotext, Minist Educ, Shanghai 201620, PR China; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, China.
| |
Collapse
|
168
|
Zhao Q, Du P, Wang X, Huang M, Sun LD, Wang T, Wang Z. Upconversion Fluorescence Resonance Energy Transfer Aptasensors for H5N1 Influenza Virus Detection. ACS OMEGA 2021; 6:15236-15245. [PMID: 34151102 PMCID: PMC8210407 DOI: 10.1021/acsomega.1c01491] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/24/2021] [Indexed: 05/27/2023]
Abstract
Influenza A virus (IAV) poses a significant threat to human health, which calls for the development of efficient detection methods. The present study constructed a fluorescence resonance energy transfer (FRET) system based on novel fluorescent probes and graphene oxide (GO) for detecting H5N1 IAV hemagglutinin (HA). Here, we synthesized small (sub-20 nm) sandwich-structured upconversion nanoparticles (UCNPs) (SWUCNPs for short) with a high energy transfer efficiency, which allows for controlling the emitter in a thin shell. The π-π stacking interaction between the aptamer and GO shortens the distance between the fluorescent probe and the receptor, thereby realizing fluorescence resonance energy transfer (FRET). When HA is present, the aptamer enables changes in their conformations and move away from GO surface. Fluorescence signals display a linear relationship between HA quantitation in the range of 0.1-15 ng mL-1 and a limit of detection (LOD) of 60.9 pg mL-1. The aptasensor was also applicable in human serum samples with a linear range from 0.2 to 12 ng mL-1 and a limit of detection of 114.7 pg mL-1. This strategy suggested the promising prospect of the aptasensor in clinical applications because of the excellent sensing performance and sensitivity. This strategy may be promising for vitro diagnostics and provides new insights into the functioning of the SWUCNPs system.
Collapse
Affiliation(s)
- Qiuzi Zhao
- School
of Life Sciences, Tianjin University, 92 Weijin Road,
Nankai District, Tianjin 300072, China
| | - Ping Du
- Beijing
National Laboratory for Molecular Sciences, State Key Laboratory of
Rare Earth Materials Chemistry and Applications, PKU-HKU Joint Laboratory
in Rare Earth Materials and Bioinorganic Chemistry, College of Chemistry
and Molecular Engineering, Peking University, 202 Chengfu Road, Haidian District, Beijing 100871, China
| | - Xiaoyong Wang
- Beijing
National Laboratory for Molecular Sciences, State Key Laboratory of
Rare Earth Materials Chemistry and Applications, PKU-HKU Joint Laboratory
in Rare Earth Materials and Bioinorganic Chemistry, College of Chemistry
and Molecular Engineering, Peking University, 202 Chengfu Road, Haidian District, Beijing 100871, China
| | - Mengqian Huang
- School
of Life Sciences, Tianjin University, 92 Weijin Road,
Nankai District, Tianjin 300072, China
| | - Ling-Dong Sun
- Beijing
National Laboratory for Molecular Sciences, State Key Laboratory of
Rare Earth Materials Chemistry and Applications, PKU-HKU Joint Laboratory
in Rare Earth Materials and Bioinorganic Chemistry, College of Chemistry
and Molecular Engineering, Peking University, 202 Chengfu Road, Haidian District, Beijing 100871, China
| | - Tao Wang
- School
of Life Sciences, Tianjin University, 92 Weijin Road,
Nankai District, Tianjin 300072, China
| | - Zhiyun Wang
- School
of Environmental Science and Engineering, 135 Yaguang Road, Jinnan District, Tianjin 300350, China
| |
Collapse
|
169
|
Wu H, Wu J, Wang H, Liu Y, Han G, Zou P. Sensitive and label-free chemiluminescence detection of malathion using exonuclease-assisted dual signal amplification and G-quadruplex/hemin DNAzyme. JOURNAL OF HAZARDOUS MATERIALS 2021; 411:124784. [PMID: 33450635 DOI: 10.1016/j.jhazmat.2020.124784] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Malathion is one of the most commonly used organophosphorus pesticides that can cause serious harm to the ecological environment and human health. Herein, we demonstrated a label-free chemiluminescent aptasensor for the sensitive detection of malathion based on exonuclease-assisted dual signal amplification and G-quadruplex/hemin DNAzyme. Upon the addition of malathion, the aptamer probe specifically bound to the target to form a complex malathion-S3, leaving a duplex S1-S2. The complex malathion-S3 was digested by exonuclease I and the target was released. The released target was recycled to perform exonuclease I-assisted signal amplification. Furthermore, after treatment with exonuclease III, the duplex S1-S2 was converted into the secondary target ST. The secondary target ST interacted with the hairpin H1 to form a complex H1-ST, which was further digested by exonuclease III and released the secondary target. The released secondary target was recycled to perform exonuclease III-assisted signal amplification. After complete amplification, large numbers of G-quadruplex/hemin DNAzymes were generated. Under the optimal experimental conditions, the prepared aptasensor showed an excellent linear response to malathion with a detection limit of 0.47 pM. The relative standard deviations were in the range of 4.2-6.9%. Moreover, the aptasensor was successfully applied to detect malathion in spiked food and traditional Chinese medicine samples.
Collapse
Affiliation(s)
- Hao Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China.
| | - Jun Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Hongyong Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yaling Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Guoqing Han
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Pei Zou
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
170
|
Roy D, Pascher A, Juratli MA, Sporn JC. The Potential of Aptamer-Mediated Liquid Biopsy for Early Detection of Cancer. Int J Mol Sci 2021; 22:ijms22115601. [PMID: 34070509 PMCID: PMC8199038 DOI: 10.3390/ijms22115601] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 01/08/2023] Open
Abstract
The early detection of cancer favors a greater chance of curative treatment and long-term survival. Exciting new technologies have been developed that can help to catch the disease early. Liquid biopsy is a promising non-invasive tool to detect cancer, even at an early stage, as well as to continuously monitor disease progression and treatment efficacy. Various methods have been implemented to isolate and purify bio-analytes in liquid biopsy specimens. Aptamers are short oligonucleotides consisting of either DNA or RNA that are capable of binding to target molecules with high specificity. Due to their unique properties, they are considered promising recognition ligands for the early detection of cancer by liquid biopsy. A variety of circulating targets have been isolated with high affinity and specificity by facile modification and affinity regulation of the aptamers. In this review, we discuss recent progress in aptamer-mediated liquid biopsy for cancer detection, its associated challenges, and its future potential for clinical applications.
Collapse
Affiliation(s)
- Dhruvajyoti Roy
- Helio Health, Irvine, CA 92618, USA
- Correspondence: ; Tel.: +1-949-8722383
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (A.P.); (M.A.J.); (J.C.S.)
| | - Mazen A. Juratli
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (A.P.); (M.A.J.); (J.C.S.)
| | - Judith C. Sporn
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (A.P.); (M.A.J.); (J.C.S.)
| |
Collapse
|
171
|
Smart Nucleic Acids as Future Therapeutics. Trends Biotechnol 2021; 39:1289-1307. [PMID: 33980422 DOI: 10.1016/j.tibtech.2021.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 11/23/2022]
Abstract
Nucleic acid therapeutics (NATs) hold promise in treating undruggable diseases and are recognized as the third major category of therapeutics in addition to small molecules and antibodies. Despite the milestones that NATs have made in clinical translation over the past decade, one important challenge pertains to increasing the specificity of this class of drugs. Activating NATs exclusively in disease-causing cells is highly desirable because it will safely broaden the application of NATs to a wider range of clinical indications. Smart NATs are triggered through a photo-uncaging reaction or a specific molecular input such as a transcript, protein, or small molecule, thus complementing the current strategy of targeting cells and tissues with receptor-specific ligands to enhance specificity. This review summarizes the programmable modalities that have been incorporated into NATs to build in responsive behaviors. We discuss the various inputs, transduction mechanisms, and output response functions that have been demonstrated to date.
Collapse
|
172
|
Li L, Wan J, Wen X, Guo Q, Jiang H, Wang J, Ren Y, Wang K. Identification of a New DNA Aptamer by Tissue-SELEX for Cancer Recognition and Imaging. Anal Chem 2021; 93:7369-7377. [PMID: 33960774 DOI: 10.1021/acs.analchem.1c01445] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer has become one of the most common diseases with high mortality in humans. Early and accurate diagnosis of cancer is of great significance to enhance the survival rate of patients. Therefore, effective molecular ligands capable of selectively recognizing cancer are urgently needed. In this work, we identified a new DNA aptamer named SW1 by tissue-based systematic evolution of ligands by exponential enrichment (tissue-SELEX), in which cancerous liver tissue sections were used as the positive control and adjacent normal liver tissue sections were used as the negative control. Taking immobilized liver cancer SMMC-7721 cells as the research object, aptamer SW1 exhibited excellent affinity with a Kd value of 123.62 ± 17.53 nM, and its binding target was preliminarily determined as a non-nucleic acid substance in the nucleus. Moreover, tissue imaging results showed that SW1 explicitly recognized cancerous liver tissues with a high detection rate of 72.7% but displayed a low detection rate to adjacent normal tissues. In addition to liver cancer cells and tissues, aptamer SW1 has been demonstrated to recognize various other types of cancer cells and tissues. Furthermore, SW1-A, an optimized aptamer of SW1, maintained its excellent affinity toward liver cancer cells and tissues. Collectively, these results indicate that SW1 possesses great potential for use as an effective molecular probe for clinical diagnosis of cancer.
Collapse
Affiliation(s)
- Lie Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| | - Jun Wan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| | - Xiaohong Wen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| | - Qiuping Guo
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| | - Huishan Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| | - Jie Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| | - Yazhou Ren
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Hunan University, Changsha 410082, China
| |
Collapse
|
173
|
Song M, Khan IM, Wang Z. Research Progress of Optical Aptasensors Based on AuNPs in Food Safety. FOOD ANAL METHOD 2021. [DOI: 10.1007/s12161-021-02029-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
174
|
Jin X, Chen L, Zhang Y, Wang X, Zhou N. A lateral flow strip for on-site detection of tobramycin based on dual-functional platinum-decorated gold nanoparticles. Analyst 2021; 146:3608-3616. [PMID: 33928957 DOI: 10.1039/d1an00403d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A novel lateral flow strip assay has been developed for rapid on-site detection of tobramycin. In this assay, unique dual-functional platinum-decorated gold nanoparticles (Au@Pt NPs) are synthesized by covering conventional gold nanoparticles (AuNPs) with an ultra-thin Pt film. Au@Pt NPs retain the plasmon activity of AuNPs and exhibit ultra-high catalytic activity that the Pt skin can achieve. The aptamer (Apt) specific for tobramycin and its complementary DNA (cDNA) are loaded on Au@Pt NPs as a duplex probe through the thiol group modified at the 5' end of the cDNA. When tobramycin is present, it binds specifically to the aptamer, resulting in its dehybridization from the cDNA and detachment from the surface of Au@Pt NPs. Then Au@Pt NPs can be captured by the fixed probe (DNA1) on the test zone (T zone) of the lateral flow strip through the hybridization between DNA1 and cDNA. The dual-functional Au@Pt NPs provide two different detection modes: one is based on the color of AuNPs (low sensitivity mode) and the other is based on the chromogenic reaction catalyzed by the Pt nanozyme (high sensitivity mode). The strip can complete the visual detection process of tobramycin within 10 min, and the cutoff values for the naked eye detection in the two modes are 60 nM and 5 nM, respectively. Furthermore, using a portable scanning reader and ImageJ software, quantitative detection can be achieved. The limits of detection (LOD) of the two modes are 0.09 nM and 0.02 nM, respectively. The strip has been successfully applied to detect tobramycin in different food samples. Therefore, Au@Pt NPs and the strip provide a highly sensitive, rapid and economical way for in-spot detection of tobramycin residues. The strip can be run in two modes, which can realize the on-demand adjustment of the detection performance and offer wider application prospects in diverse scenarios.
Collapse
Affiliation(s)
- Xin Jin
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Luo Chen
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Yuting Zhang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Xiaoli Wang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Nandi Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
175
|
Thavarajah W, Hertz LM, Bushhouse DZ, Archuleta CM, Lucks JB. RNA Engineering for Public Health: Innovations in RNA-Based Diagnostics and Therapeutics. Annu Rev Chem Biomol Eng 2021; 12:263-286. [PMID: 33900805 PMCID: PMC9714562 DOI: 10.1146/annurev-chembioeng-101420-014055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
RNA is essential for cellular function: From sensing intra- and extracellular signals to controlling gene expression, RNA mediates a diverse and expansive list of molecular processes. A long-standing goal of synthetic biology has been to develop RNA engineering principles that can be used to harness and reprogram these RNA-mediated processes to engineer biological systems to solve pressing global challenges. Recent advances in the field of RNA engineering are bringing this to fruition, enabling the creation of RNA-based tools to combat some of the most urgent public health crises. Specifically, new diagnostics using engineered RNAs are able to detect both pathogens and chemicals while generating an easily detectable fluorescent signal as an indicator. New classes of vaccines and therapeutics are also using engineered RNAs to target a wide range of genetic and pathogenic diseases. Here, we discuss the recent breakthroughs in RNA engineering enabling these innovations and examine how advances in RNA design promise to accelerate the impact of engineered RNA systems.
Collapse
Affiliation(s)
- Walter Thavarajah
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, USA; .,Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, USA.,Center for Water Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Laura M Hertz
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, USA.,Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois 60208, USA
| | - David Z Bushhouse
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, USA.,Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois 60208, USA
| | - Chloé M Archuleta
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, USA; .,Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, USA.,Center for Water Research, Northwestern University, Evanston, Illinois 60208, USA
| | - Julius B Lucks
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, USA; .,Center for Synthetic Biology, Northwestern University, Evanston, Illinois 60208, USA.,Center for Water Research, Northwestern University, Evanston, Illinois 60208, USA.,Center for Engineering Sustainability and Resilience, Northwestern University, Evanston, Illinois 60208, USA
| |
Collapse
|
176
|
Yi K, Rong Y, Huang L, Tang X, Zhang Q, Wang W, Wu J, Wang F. Aptamer-Exosomes for Tumor Theranostics. ACS Sens 2021; 6:1418-1429. [PMID: 33755415 DOI: 10.1021/acssensors.0c02237] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
As carriers of biomolecules (proteins, nucleic acids, and lipids) from parent cells, exosomes play a significant role in physiology and pathology. In any diseased state, the morphology of the released exosomes remained similar. The contents of exosomes change depending on the disease or its stage; thus, exosomes are generally considered as a "source of biomarkers". Therefore, they are considered promising biomarkers for the diagnosis and prognosis of tumors. As natural delivery vehicles, exosomes can protect their cargo from immune clearance and deliver them to other cells through membrane fusion. After being genetically edited at the cell or exosome level, exosomes can be used for treatment with aptamers. Aptamers are short stretches of oligonucleotide sequences or short polypeptides that have been selected in vitro or in vivo, and have a wide range of targets and show excellent binding affinity and specificity. Aptamers have been widely used as molecular probes, and the combination of aptamers with exosomes has become a new direction for exosome-related research and therapeutic development. Here, we summarized various applications of exosomes and aptamers in cancer research, and further analyzed their combination as an "aptamer-exosome". Finally, we propose future directions for the aptamer-exosome in the precise diagnosis or personalized treatment of cancer.
Collapse
Affiliation(s)
- Kezhen Yi
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| | - Yuan Rong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| | - Lanxiang Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| | - Xuan Tang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| | - Qian Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| | - Wei Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| | - Jianyuan Wu
- Clinical Trial Center of Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, No. 169 Donghu Road, Wuchang District, Wuhan 430071, P.R. China
| |
Collapse
|
177
|
Kvízová J, Pavlíčková V, Kmoníčková E, Ruml T, Rimpelová S. Quo Vadis Advanced Prostate Cancer Therapy? Novel Treatment Perspectives and Possible Future Directions. Molecules 2021; 26:2228. [PMID: 33921501 PMCID: PMC8069564 DOI: 10.3390/molecules26082228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022] Open
Abstract
Prostate cancer is a very common disease, which is, unfortunately, often the cause of many male deaths. This is underlined by the fact that the early stages of prostate cancer are often asymptomatic. Therefore, the disease is usually detected and diagnosed at late advanced or even metastasized stages, which are already difficult to treat. Hence, it is important to pursue research and development not only in terms of novel diagnostic methods but also of therapeutic ones, as well as to increase the effectiveness of the treatment by combinational medicinal approach. Therefore, in this review article, we focus on recent approaches and novel potential tools for the treatment of advanced prostate cancer; these include not only androgen deprivation therapy, antiandrogen therapy, photodynamic therapy, photothermal therapy, immunotherapy, multimodal therapy, but also poly(ADP-ribose) polymerase, Akt and cyclin-dependent kinase inhibitors.
Collapse
Affiliation(s)
- Jana Kvízová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
- Bioinova, s.r.o., Vídeňská 1083, 140 20 Praha, Czech Republic
| | - Vladimíra Pavlíčková
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
| | - Eva Kmoníčková
- Institute of Experimental Medicine of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic;
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague, Czech Republic; (J.K.); (V.P.); (T.R.)
| |
Collapse
|
178
|
Ștefan G, Hosu O, De Wael K, Lobo-Castañón MJ, Cristea C. Aptamers in biomedicine: Selection strategies and recent advances. Electrochim Acta 2021. [DOI: 10.1016/j.electacta.2021.137994] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
179
|
BipD of Burkholderia pseudomallei: Structure, Functions, and Detection Methods. Microorganisms 2021; 9:microorganisms9040711. [PMID: 33808203 PMCID: PMC8067316 DOI: 10.3390/microorganisms9040711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 01/13/2023] Open
Abstract
Melioidosis is a severe disease caused by Burkholderia pseudomallei (B. pseudomallei), a Gram-negative environmental bacterium. It is endemic in Southeast Asia and Northern Australia, but it is underreported in many other countries. The principal routes of entry for B. pseudomallei are skin penetration, inhalation, and ingestion. It mainly affects immunocompromised populations, especially patients with type 2 diabetes mellitus. The laboratory diagnosis of melioidosis is challenging due to its non-specific clinical manifestations, which mimic other severe infections. The culture method is considered an imperfect gold standard for the diagnosis of melioidosis due to its low sensitivity. Antibody detection has low sensitivity and specificity due to the high seropositivity among healthy people in endemic regions. Antigen detection using various proteins has been tested for the rapid determination of B. pseudomallei; however, it presents certain limitations in terms of its sensitivity and specificity. Therefore, this review aims to frame the present knowledge of a potential target known as the Burkholderia invasion protein D (BipD), including future directions for its detection using an aptamer-based sensor (aptasensor).
Collapse
|
180
|
The use of aptamers in prostate cancer: A systematic review of theranostic applications. Clin Biochem 2021; 93:9-25. [PMID: 33794195 DOI: 10.1016/j.clinbiochem.2021.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
Since prostate cancer (PCa) relies on limited diagnosis and therapies, more effective alternatives are needed. Aptamers are versatile tools that may be applied for better clinical management of PCa patients. This review shows the trends on aptamer-based applications for PCa to understand their future development. We searched articles reporting aptamers applied in PCa on the Pubmed, Scopus and Web of Science databases over the last decade. Almost 80% of the articles used previously selected aptamers in novel approaches. However, cell-SELEX was the most applied technique for the selection of new aptamers allowing their binding to targets in their native configuration. ssDNA aptamers were 24% more common than RNA aptamers. The most studied PCa-specific aptamers were the DNA PSA-specific aptamer PSap4#5 and the PSMA-specific RNA aptamers A10 and A9, being PSA and PSMA the most reported targets. Thus, researchers still prefer the ease of use of DNA aptamers. Blood-based liquid biopsies represented 24% of all samples, being the most promising clinical samples. Especially noteworthy, electro-analytical methods accounted for more than 40% of the diagnostic techniques and treatment approaches with drug delivery systems or transcriptional modifiers were reported in 70% of the articles. Although all these articles showed clinically relevant aptamers for PCa and there are good prospects for their use, the development of all these strategies was in its early stages. Thus, the aptamers are not completely validated and we foresee that the completion of clinical studies will allow the implementation of these aptamer-based technologies in the clinical practice of PCa.
Collapse
|
181
|
Gao T, Mao Z, Li W, Pei R. Anti-PD-L1 DNA aptamer antagonizes the interaction of PD-1/PD-L1 with antitumor effect. J Mater Chem B 2021; 9:746-756. [PMID: 33319876 DOI: 10.1039/d0tb01668c] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor immune evasion enables cancer cells to escape destruction by the immune system, which causes poor prognosis and overall survival of some tumor patients. The binding of PD-L1 on tumor cells to PD-1 on T cells suppresses T cell function, and the axis is considered one of the major pathways mediating tumor cells to evade immune surveillance. The PD-L1 ligation of T cells has a profound inhibitory effect on the growth, cytokine secretion, and development of cytotoxicity. Aptamers, known as chemical antibodies, are single-stranded oligonucleotides with high affinity. In this work, we take a cell-SELEX with the engineered PD-L1-expressing cells as a target to obtain the aptamer, designated PL1, which specifically binds to PD-L1 with a Kd value of 95.73 nM, resulting in the inhibition of PD-1/PD-L1. The aptamer PL1 could restore the proliferation and IFN-γ rescue from the T cell inhibited by the PD-1/PD-L1 axis, and inhibit the growth of the CT26 colon carcinoma. The similar tumor inhibition efficacy and binding capacity of the aptamer PL1 as an antibody indicate that the aptamer PL1 can serve as an alternative therapeutic agent for cancer immunotherapy since the use of antibodies is often restricted by high cost, large size and poor tumor penetration.
Collapse
Affiliation(s)
- Tian Gao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | | | | | | |
Collapse
|
182
|
Li J, Ren X, Zhao J, Lou X. PD-L1 aptamer isolation via Modular-SELEX and its applications in cancer cell detection and tumor tissue section imaging. Analyst 2021; 146:2910-2918. [PMID: 33724284 DOI: 10.1039/d1an00182e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PD-1/PD-L1 is an important pathway in immunotherapy and a high PD-L1 expression level in tumor tissues is an essential prerequisite for PD-1/PD-L1 blocking-based therapy. The PD-L1 expression level in tumor tissue sections is currently detected via immunohistochemistry (IHC) using anti-PD-L1 antibodies from various resources, which has the disadvantage of inconsistent results. As synthetic affinity ligands, aptamers have good batch-to-batch consistency and have been demonstrated to have great potential for use in biomedical applications. In this study, we isolated PD-L1 aptamers using a combination method, named Modular-SELEX (systematic evolution of ligands by exponential enrichment), which includes three sequentially performed modules: the affinity module, the specificity module, and the compatibility module. Three rounds of magnetic crosslinking precipitation (MCP)-SELEX, three rounds of Capture-SELEX, and two rounds of Tissue-SELEX were respectively performed in the corresponding three modules to significantly and efficiently improve the native affinity, specificity, and compatibility of the enriched library. The isolated aptamer Clon-3 had nanomolar binding affinity, as determined via both homogeneous and PD-L1 immobilized affinity assays. Clon-3 could be used to recognize various cancer cells with distinct PD-L1 expression levels using flow cytometry. The PD-L1 expression levels in normal human tonsils (the gold standard for anti-PD-L1 antibody) and non-small cell lung cancer tissue sections stained using Cy5.5-labeled Clon-3 were also successfully imaged using a confocal microscope. The fluorescence intensities of the tissue sections were in good agreement with their actual PD-L1 expression levels as confirmed via IHC.
Collapse
Affiliation(s)
- Jiyuan Li
- Department of Chemistry, Capital Normal University, Xisanhuan North Road 105, Beijing 100048, China.
| | - Xijiao Ren
- Department of Chemistry, Capital Normal University, Xisanhuan North Road 105, Beijing 100048, China.
| | - Jiaxing Zhao
- Department of Chemistry, Capital Normal University, Xisanhuan North Road 105, Beijing 100048, China.
| | - Xinhui Lou
- Department of Chemistry, Capital Normal University, Xisanhuan North Road 105, Beijing 100048, China.
| |
Collapse
|
183
|
Shanaa OA, Rumyantsev A, Sambuk E, Padkina M. In Vivo Production of RNA Aptamers and Nanoparticles: Problems and Prospects. Molecules 2021; 26:molecules26051422. [PMID: 33800717 PMCID: PMC7961669 DOI: 10.3390/molecules26051422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/26/2022] Open
Abstract
RNA aptamers are becoming increasingly attractive due to their superior properties. This review discusses the early stages of aptamer research, the main developments in this area, and the latest technologies being developed. The review also highlights the advantages of RNA aptamers in comparison to antibodies, considering the great potential of RNA aptamers and their applications in the near future. In addition, it is shown how RNA aptamers can form endless 3-D structures, giving rise to various structural and functional possibilities. Special attention is paid to the Mango, Spinach and Broccoli fluorescent RNA aptamers, and the advantages of split RNA aptamers are discussed. The review focuses on the importance of creating a platform for the synthesis of RNA nanoparticles in vivo and examines yeast, namely Saccharomyces cerevisiae, as a potential model organism for the production of RNA nanoparticles on a large scale.
Collapse
Affiliation(s)
- Ousama Al Shanaa
- Department of Genetics and Biotechnology, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.R.); (E.S.)
- Atomic Energy Commission of Syria, Damascus P.O.B 6091, Syria
- Correspondence: (O.A.S.); (M.P.); Tel.: +7-812-328-2822 (O.A.S.); +7-812-327-9827 (M.P.)
| | - Andrey Rumyantsev
- Department of Genetics and Biotechnology, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.R.); (E.S.)
| | - Elena Sambuk
- Department of Genetics and Biotechnology, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.R.); (E.S.)
| | - Marina Padkina
- Department of Genetics and Biotechnology, Saint Petersburg State University, 199034 Saint Petersburg, Russia; (A.R.); (E.S.)
- Correspondence: (O.A.S.); (M.P.); Tel.: +7-812-328-2822 (O.A.S.); +7-812-327-9827 (M.P.)
| |
Collapse
|
184
|
Lyu C, Khan IM, Wang Z. Capture-SELEX for aptamer selection: A short review. Talanta 2021; 229:122274. [PMID: 33838776 DOI: 10.1016/j.talanta.2021.122274] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022]
Abstract
The emerging aptamer, developed through the systematic evolution of ligands by exponential enrichment (SELEX) process, has revolutionized and facilitated the discoveries in basic research. Among all SELEX technology, Capture-SELEX is a variant of the in vitro selection process, which is suitable for isolating aptamers against small molecules. Capture-SELEX library was developed to enable the immobilization of the oligonucleotides instead of the target molecules during the aptamer selection process. The review provides an update on the recent-advances in this new screening method with particular emphasis on key points of capture protocol and its applications. The limitations and the prospects of the Capture-SELEX are also discussed. We hope that present review will inspire more researchers to understand the selection problems from the perspective of Capture-SELEX. Moreover, it will open new pave to improve the efficiency and success of screening to meet the growing demand for aptasensor discovery in small molecules.
Collapse
Affiliation(s)
- Chen Lyu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Imran Mahmood Khan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China; Synergetic Innovation Center of Food Safety and Quality Control of Jiangsu Province, China
| | - Zhouping Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China; Synergetic Innovation Center of Food Safety and Quality Control of Jiangsu Province, China.
| |
Collapse
|
185
|
Li J, Gu J, Zhang H, Liu R, Zhang W, Mohammed-Elsabagh M, Xia J, Morrison D, Zakaria S, Chang D, Arrabi A, Li Y. A Highly Specific DNA Aptamer for RNase H2 from Clostridium difficile. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9464-9471. [PMID: 33410654 DOI: 10.1021/acsami.0c20277] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Molecular recognition elements with high specificity are of great importance for the study of molecular interactions, accurate diagnostics, drug design, and personalized medicine. Herein, a highly specific DNA aptamer for RNase H2 from Clostridium difficile (C. difficile) was generated by SELEX and minimized to 40 nucleotides. The aptamer exhibits a dissociation constant (Kd) of 1.8 ± 0.5 nM and an inhibition constant (IC50) of 7.1 ± 0.6 nM for C. difficile RNase H2, both of which are 2 orders of magnitude better for the same enzyme from other control bacteria. The fluorescent version of the aptamer can distinguish C. difficile from several other control bacteria in a cell lysate assay. This work demonstrates that a ubiquitous protein like RNase H2 can still be used as the target for the development of highly specific aptamers and the combination of the protein and the aptamer can achieve the recognition specificity needed for a diagnostic test and drug development.
Collapse
Affiliation(s)
- Jiuxing Li
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Jimmy Gu
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Hongfen Zhang
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Rudi Liu
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Wenqing Zhang
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Mostafa Mohammed-Elsabagh
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Jianrun Xia
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Devon Morrison
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Sandy Zakaria
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Dingran Chang
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Amjad Arrabi
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| | - Yingfu Li
- M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
186
|
Sun M, Chen Z, Wu X, Yu Y, Wang L, Lu A, Zhang G, Li F. The Roles of Sclerostin in Immune System and the Applications of Aptamers in Immune-Related Research. Front Immunol 2021; 12:602330. [PMID: 33717084 PMCID: PMC7946814 DOI: 10.3389/fimmu.2021.602330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/14/2021] [Indexed: 12/19/2022] Open
Abstract
Wnt signaling is one of the fundamental pathways that play a major role in almost every aspect of biological systems. In addition to the well-known influence of Wnt signaling on bone formation, its essential role in the immune system also attracted increasing attention. Sclerostin, a confirmed Wnt antagonist, is also proven to modulate the development and differentiation of normal immune cells, particularly B cells. Aptamers, single-stranded (ss) oligonucleotides, are capable of specifically binding to a variety of target molecules by virtue of their unique three-dimensional structures. With in-depth study of those functional nucleic acids, they have been gradually applied to diagnostic and therapeutic area in immune diseases due to their various advantages over antibodies. In this review, we focus on several issues including the roles of Wnt signaling and Wnt antagonist sclerostin in the immune system. For the sake of understanding, current examples of aptamers applications for the immune diseases are also discussed. At the end of this review, we propose our ideas for the future research directions.
Collapse
Affiliation(s)
- Meiheng Sun
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoqiu Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| |
Collapse
|
187
|
Saad M, Faucher SP. Aptamers and Aptamer-Coupled Biosensors to Detect Water-Borne Pathogens. Front Microbiol 2021; 12:643797. [PMID: 33679681 PMCID: PMC7933031 DOI: 10.3389/fmicb.2021.643797] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Aptamers can serve as efficient bioreceptors for the development of biosensing detection platforms. Aptamers are short DNA or RNA oligonucleotides that fold into specific structures, which enable them to selectively bind to target analytes. The method used to identify aptamers is Systematic Evolution of Ligands through Exponential Enrichment (SELEX). Target properties can have an impact on aptamer efficiencies. Therefore, characteristics of water-borne microbial targets must be carefully considered during SELEX for optimal aptamer development. Several aptamers have been described for key water-borne pathogens. Here, we provide an exhaustive overview of these aptamers and discuss important microbial aspects to consider when developing such aptamers.
Collapse
Affiliation(s)
- Mariam Saad
- Department of Natural Resources, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, QC, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, Faculté de Médecine Vétérinaire, Saint-Hyacinthe, QC, Canada
| | - Sebastien P. Faucher
- Department of Natural Resources, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, QC, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, Faculté de Médecine Vétérinaire, Saint-Hyacinthe, QC, Canada
| |
Collapse
|
188
|
Zhao X, Dai X, Zhao S, Cui X, Gong T, Song Z, Meng H, Zhang X, Yu B. Aptamer-based fluorescent sensors for the detection of cancer biomarkers. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 247:119038. [PMID: 33120124 DOI: 10.1016/j.saa.2020.119038] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
Aptamers are short single-stranded RNA or DNA molecules that can recognize a series of targets with high affinity and specificity. Known as "chemical antibodies", aptamers have many unique merits, including ease of chemical synthesis, high chemical stability, low molecular weight, lack of immunogenicity, and ease of modification and manipulation compared to their protein counterparts. Using aptamers as the recognition groups, fluorescent aptasensors provide exciting opportunities for sensitive detection and quantification of analytes. Herein, we give an overview on the recent development of aptamer-based fluorescent sensors for the detection of cancer biomarkers. Based on various nanostructured sensor designs, we extended our discussions on sensitivity, specificity and the potential applications of aptamer-based fluorescent sensors in early diagnosis, treatment and prognosis of cancers.
Collapse
Affiliation(s)
- Xuhua Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaochun Dai
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Suya Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaohua Cui
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Tao Gong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhiling Song
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Hongmin Meng
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaobing Zhang
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Baofeng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
189
|
Moretta R, De Stefano L, Terracciano M, Rea I. Porous Silicon Optical Devices: Recent Advances in Biosensing Applications. SENSORS (BASEL, SWITZERLAND) 2021; 21:1336. [PMID: 33668616 PMCID: PMC7917735 DOI: 10.3390/s21041336] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023]
Abstract
This review summarizes the leading advancements in porous silicon (PSi) optical-biosensors, achieved over the past five years. The cost-effective fabrication process, the high internal surface area, the tunable pore size, and the photonic properties made the PSi an appealing transducing substrate for biosensing purposes, with applications in different research fields. Different optical PSi biosensors are reviewed and classified into four classes, based on the different biorecognition elements immobilized on the surface of the transducing material. The PL signal modulation and the effective refractive index changes of the porous matrix are the main optical transduction mechanisms discussed herein. The approaches that are commonly employed to chemically stabilize and functionalize the PSi surface are described.
Collapse
Affiliation(s)
- Rosalba Moretta
- National Research Council, Institute of Applied Sciences and Intelligent Systems, Unit of Naples, 80131 Naples, Italy; (R.M.); (L.D.S.); (I.R.)
| | - Luca De Stefano
- National Research Council, Institute of Applied Sciences and Intelligent Systems, Unit of Naples, 80131 Naples, Italy; (R.M.); (L.D.S.); (I.R.)
| | - Monica Terracciano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Ilaria Rea
- National Research Council, Institute of Applied Sciences and Intelligent Systems, Unit of Naples, 80131 Naples, Italy; (R.M.); (L.D.S.); (I.R.)
| |
Collapse
|
190
|
Shi X, Chen M, Feng H, Zhou Z, Wu R, Li W, Liang J, Chen J, Li G. Glypican-3 electrochemical aptasensor based on reduced graphene oxide‐chitosan‐ferrocene deposition of platinum–palladium bimetallic nanoparticles. J APPL ELECTROCHEM 2021. [DOI: 10.1007/s10800-021-01534-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
191
|
Vázquez-González M, Willner I. Aptamer-Functionalized Hybrid Nanostructures for Sensing, Drug Delivery, Catalysis and Mechanical Applications. Int J Mol Sci 2021; 22:1803. [PMID: 33670386 PMCID: PMC7918352 DOI: 10.3390/ijms22041803] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 01/05/2023] Open
Abstract
Sequence-specific nucleic acids exhibiting selective recognition properties towards low-molecular-weight substrates and macromolecules (aptamers) find growing interest as functional biopolymers for analysis, medical applications such as imaging, drug delivery and even therapeutic agents, nanotechnology, material science and more. The present perspective article introduces a glossary of examples for diverse applications of aptamers mainly originated from our laboratory. These include the introduction of aptamer-functionalized nanomaterials such as graphene oxide, Ag nanoclusters and semiconductor quantum dots as functional hybrid nanomaterials for optical sensing of target analytes. The use of aptamer-functionalized DNA tetrahedra nanostructures for multiplex analysis and aptamer-loaded metal-organic framework nanoparticles acting as sense-and-treat are introduced. Aptamer-functionalized nano and microcarriers are presented as stimuli-responsive hybrid drug carriers for controlled and targeted drug release, including aptamer-functionalized SiO2 nanoparticles, carbon dots, metal-organic frameworks and microcapsules. A further application of aptamers involves the conjugation of aptamers to catalytic units as a means to mimic enzyme functions "nucleoapzymes". In addition, the formation and dissociation of aptamer-ligand complexes are applied to develop mechanical molecular devices and to switch nanostructures such as origami scaffolds. Finally, the article discusses future challenges in applying aptamers in material science, nanotechnology and catalysis.
Collapse
Affiliation(s)
- Margarita Vázquez-González
- Center for Nanoscience and Nanotechnology, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Itamar Willner
- Center for Nanoscience and Nanotechnology, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
192
|
Tompkins KJ, Houtti M, Litzau LA, Aird EJ, Everett BA, Nelson AT, Pornschloegl L, Limón-Swanson LK, Evans RL, Evans K, Shi K, Aihara H, Gordon WR. Molecular underpinnings of ssDNA specificity by Rep HUH-endonucleases and implications for HUH-tag multiplexing and engineering. Nucleic Acids Res 2021; 49:1046-1064. [PMID: 33410911 PMCID: PMC7826260 DOI: 10.1093/nar/gkaa1248] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Replication initiator proteins (Reps) from the HUH-endonuclease superfamily process specific single-stranded DNA (ssDNA) sequences to initiate rolling circle/hairpin replication in viruses, such as crop ravaging geminiviruses and human disease causing parvoviruses. In biotechnology contexts, Reps are the basis for HUH-tag bioconjugation and a critical adeno-associated virus genome integration tool. We solved the first co-crystal structures of Reps complexed to ssDNA, revealing a key motif for conferring sequence specificity and for anchoring a bent DNA architecture. In combination, we developed a deep sequencing cleavage assay, termed HUH-seq, to interrogate subtleties in Rep specificity and demonstrate how differences can be exploited for multiplexed HUH-tagging. Together, our insights allowed engineering of only four amino acids in a Rep chimera to predictably alter sequence specificity. These results have important implications for modulating viral infections, developing Rep-based genomic integration tools, and enabling massively parallel HUH-tag barcoding and bioconjugation applications.
Collapse
Affiliation(s)
- Kassidy J Tompkins
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Mo Houtti
- Department of Computer Science and Engineering, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Lauren A Litzau
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Eric J Aird
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Blake A Everett
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Andrew T Nelson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Leland Pornschloegl
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Lidia K Limón-Swanson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Robert L Evans
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Karen Evans
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Wendy R Gordon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
193
|
Herkt M, Thum T. Pharmacokinetics and Proceedings in Clinical Application of Nucleic Acid Therapeutics. Mol Ther 2021; 29:521-539. [PMID: 33188937 PMCID: PMC7854291 DOI: 10.1016/j.ymthe.2020.11.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/27/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
Oligonucleotide therapeutics are a novel promising class of drugs designed to specifically target either coding or non-coding RNA molecules to revolutionize treatment of various diseases. During preclinical development, investigations of the pharmacokinetic characteristics of these oligonucleotide-based drug candidates are essential. Oligonucleotides possess a long history of chemical modifications to enhance their stability and binding affinity, as well as reducing toxicity. Phosphorothioate backbone modifications of oligonucleotides were a hallmark of this development process that greatly enhanced plasma stability and protein binding of these agents. Modifications such as 2'-O-methylation further improved stability, while other modifications of the ribose, such as locked nucleic acid (LNA) modification, significantly increased binding affinity, potency, and tissue half-life. These attributes render oligonucleotide therapeutics able to regulate protein expression in both directions depending on the target RNA. Thus, a growing interest has emerged using these oligonucleotides in the treatment of neurodegenerative and cardiac disorders as well as cancer, since the deregulation of certain coding and non-coding RNAs plays a key role in the development of these diseases. Cutting edge research is being performed in the field of non-coding RNAs, identifying potential therapeutic targets, and developing novel oligonucleotide-based agents that outperform classical drugs. Some of these agents are either in clinical trials showing promising results or are already US Food and Drug Administration (FDA) approved, with more oligonucleotides being developed for therapeutic purposes. This is the advent of mechanism-based next-generation therapeutics for a wide range of diseases.
Collapse
Affiliation(s)
- Markus Herkt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School (MHH), Hannover, Germany.
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School (MHH), Hannover, Germany; REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School (MHH), Hannover, Germany; Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany.
| |
Collapse
|
194
|
|
195
|
Chudinov AV, Vasiliskov VA, Kuznetsova VE, Lapa SA, Kolganova NA, Timofeev EN. Mononucleotide repeat expansions with non-natural polymerase substrates. Sci Rep 2021; 11:2423. [PMID: 33510377 PMCID: PMC7844250 DOI: 10.1038/s41598-021-82150-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Replicative strand slippage is a biological phenomenon, ubiquitous among different organisms. However, slippage events are also relevant to non-natural replication models utilizing synthetic polymerase substrates. Strand slippage may notably affect the outcome of the primer extension reaction with repetitive templates in the presence of non-natural nucleoside triphosphates. In the current paper, we studied the ability of Taq, Vent (exo-), and Deep Vent (exo-) polymerases to produce truncated, full size, or expanded modified strands utilizing non-natural 2′-deoxyuridine nucleotide analogues and different variants of the homopolymer template. Our data suggest that the slippage of the primer strand is dependent on the duplex fluttering, incorporation efficiency for a particular polymerase-dNTP pair, rate of non-templated base addition, and presence of competing nucleotides.
Collapse
Affiliation(s)
- Alexander V Chudinov
- W. A. Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov St. 32, Moscow, Russia, 119991
| | - Vadim A Vasiliskov
- W. A. Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov St. 32, Moscow, Russia, 119991
| | - Viktoriya E Kuznetsova
- W. A. Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov St. 32, Moscow, Russia, 119991
| | - Sergey A Lapa
- W. A. Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov St. 32, Moscow, Russia, 119991
| | - Natalia A Kolganova
- W. A. Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov St. 32, Moscow, Russia, 119991
| | - Edward N Timofeev
- W. A. Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Vavilov St. 32, Moscow, Russia, 119991.
| |
Collapse
|
196
|
Liu L, Gao H, Guo C, Liu T, Li N, Qian Q. Therapeutic Mechanism of Nucleic Acid Drugs. ChemistrySelect 2021. [DOI: 10.1002/slct.202002901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lianxiao Liu
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Haixia Gao
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Chuanxin Guo
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Tao Liu
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Ning Li
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| | - Qijun Qian
- Nucleic Acid Drug Division Shanghai Cell Therapy Group Co., Ltd. 75 A Qianyang Rd, Jiading District Shanghai 201805 China
| |
Collapse
|
197
|
Tadesse LF, Safir F, Ho CS, Hasbach X, Khuri-Yakub BP, Jeffrey SS, Saleh AAE, Dionne J. Toward rapid infectious disease diagnosis with advances in surface-enhanced Raman spectroscopy. J Chem Phys 2021; 152:240902. [PMID: 32610995 DOI: 10.1063/1.5142767] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In a pandemic era, rapid infectious disease diagnosis is essential. Surface-enhanced Raman spectroscopy (SERS) promises sensitive and specific diagnosis including rapid point-of-care detection and drug susceptibility testing. SERS utilizes inelastic light scattering arising from the interaction of incident photons with molecular vibrations, enhanced by orders of magnitude with resonant metallic or dielectric nanostructures. While SERS provides a spectral fingerprint of the sample, clinical translation is lagged due to challenges in consistency of spectral enhancement, complexity in spectral interpretation, insufficient specificity and sensitivity, and inefficient workflow from patient sample collection to spectral acquisition. Here, we highlight the recent, complementary advances that address these shortcomings, including (1) design of label-free SERS substrates and data processing algorithms that improve spectral signal and interpretability, essential for broad pathogen screening assays; (2) development of new capture and affinity agents, such as aptamers and polymers, critical for determining the presence or absence of particular pathogens; and (3) microfluidic and bioprinting platforms for efficient clinical sample processing. We also describe the development of low-cost, point-of-care, optical SERS hardware. Our paper focuses on SERS for viral and bacterial detection, in hopes of accelerating infectious disease diagnosis, monitoring, and vaccine development. With advances in SERS substrates, machine learning, and microfluidics and bioprinting, the specificity, sensitivity, and speed of SERS can be readily translated from laboratory bench to patient bedside, accelerating point-of-care diagnosis, personalized medicine, and precision health.
Collapse
Affiliation(s)
- Loza F Tadesse
- Department of Bioengineering, Stanford University School of Medicine and School of Engineering, Stanford, California 94305, USA
| | - Fareeha Safir
- Department of Mechanical Engineering, Stanford University School of Engineering, Stanford, California 94305, USA
| | - Chi-Sing Ho
- Department of Applied Physics, Stanford University School of Humanities and Sciences, Stanford, California 94305, USA
| | - Ximena Hasbach
- Department of Materials Science and Engineering, Stanford University School of Engineering, Stanford, California 94305, USA
| | - Butrus Pierre Khuri-Yakub
- Department of Electrical Engineering, Stanford University School of Engineering, Stanford, California 94305, USA
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Amr A E Saleh
- Department of Materials Science and Engineering, Stanford University School of Engineering, Stanford, California 94305, USA
| | - Jennifer Dionne
- Department of Materials Science and Engineering, Stanford University School of Engineering, Stanford, California 94305, USA
| |
Collapse
|
198
|
Ziółkowski R, Jarczewska M, Górski Ł, Malinowska E. From Small Molecules Toward Whole Cells Detection: Application of Electrochemical Aptasensors in Modern Medical Diagnostics. SENSORS (BASEL, SWITZERLAND) 2021; 21:724. [PMID: 33494499 PMCID: PMC7866209 DOI: 10.3390/s21030724] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
Abstract
This paper focuses on the current state of art as well as on future trends in electrochemical aptasensors application in medical diagnostics. The origin of aptamers is presented along with the description of the process known as SELEX. This is followed by the description of the broad spectrum of aptamer-based sensors for the electrochemical detection of various diagnostically relevant analytes, including metal cations, abused drugs, neurotransmitters, cancer, cardiac and coagulation biomarkers, circulating tumor cells, and viruses. We described also possible future perspectives of aptasensors development. This concerns (i) the approaches to lowering the detection limit and improvement of the electrochemical aptasensors selectivity by application of the hybrid aptamer-antibody receptor layers and/or nanomaterials; and (ii) electrochemical aptasensors integration with more advanced microfluidic devices as user-friendly medical instruments for medical diagnostic of the future.
Collapse
Affiliation(s)
- Robert Ziółkowski
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (M.J.); (Ł.G.)
| | - Marta Jarczewska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (M.J.); (Ł.G.)
| | - Łukasz Górski
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (M.J.); (Ł.G.)
| | - Elżbieta Malinowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (M.J.); (Ł.G.)
- Center for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| |
Collapse
|
199
|
Zhao X, Li S, Liu G, Wang Z, Yang Z, Zhang Q, Liang M, Liu J, Li Z, Tong Y, Zhu G, Wang X, Jiang L, Wang W, Tan GY, Zhang L. A versatile biosensing platform coupling CRISPR-Cas12a and aptamers for detection of diverse analytes. Sci Bull (Beijing) 2021; 66:69-77. [PMID: 36654316 DOI: 10.1016/j.scib.2020.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/07/2020] [Accepted: 08/22/2020] [Indexed: 01/20/2023]
Abstract
Rapid and sensitive detection of various analytes is in high demand. Apart from its application in genome editing, CRISPR-Cas also shows promises in nucleic acid detection applications. To further exploit the potential of CRISPR-Cas for detection of diverse analytes, we present a versatile biosensing platform that couples the excellent affinity of aptamers for broad-range analytes with the collateral single-strand DNA cleavage activity of CRISPR-Cas12a. We demonstrated that the biosensors developed by this platform can be used to detect protein and small molecule in human serum with a complicated background, i.e., the tumor marker alpha fetoprotein and cocaine with the detection limits of 0.07 fmol/L and 0.34 μmol/L, respectively, highlighting the advantages of simplicity, sensitivity, short detection time, and low cost compared with the state-of-the-art biosensing approaches. Altogether, this biosensing platform with plug-and-play design show great potential in the detection of diverse analytes.
Collapse
Affiliation(s)
- Xiangxiang Zhao
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China
| | - Shanshan Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guang Liu
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China
| | - Zhong Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Department of Urology, Sixth People's Hospital South Campus Affiliated to Shanghai Jiao Tong University, Shanghai 201499, China
| | - Zhiheng Yang
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China
| | - Quanwei Zhang
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China; State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Mindong Liang
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China
| | - Jiakun Liu
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zilong Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yaojun Tong
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs Lyngby 2800, Denmark
| | - Guoliang Zhu
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China
| | - Xinye Wang
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China
| | - Lan Jiang
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China
| | - Weishan Wang
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China; State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Gao-Yi Tan
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China.
| | - Lixin Zhang
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST), Shanghai 200237, China.
| |
Collapse
|
200
|
|