Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2018 May 7;77(2):83–98. doi: 10.1016/j.jinf.2018.04.007

Oligonucleotide aptamers: promising and powerful diagnostic and therapeutic tools for infectious diseases

Qin Pan 1, Fengling Luo 1, Min Liu 1, Xiao-Lian Zhang 1,
PMCID: PMC7112547  PMID: 29746951

Summary

The entire human population is at risk of infectious diseases worldwide. Thus far, the diagnosis and treatment of human infectious diseases at the molecular and nanoscale levels have been extremely challenging tasks because of the lack of effective probes to identify and recognize biomarkers of pathogens. Oligonucleotide aptamers are a class of small nucleic acid ligands that are composed of single-stranded DNA (ssDNA) or RNA and act as affinity probes or molecular recognition elements for a variety of targets. These aptamers have an exciting potential for diagnose and/or treatment of specific diseases. In this review, we highlight areas where aptamers have been developed as diagnostic and therapeutic agents for both bacterial and viral infectious diseases as well as aptamer-based detection.

Keywords: SELEX, Aptamer, Infectious disease, Diagnosis, Therapy

Introduction

Infections with a pandemic potential are major global risks, and infectious diseases in general are an important threat to public health. Preventing and controlling infectious disease are still associated with multiple challenges and obstacles. The increasing population and complexity of infectious disease are leading to synergistic effects. Novel pharmacological agents need to be developed for rapid diagnosis and cutting off emerging infectious diseases at the source. Exploration of novel molecular recognition elements specific for pathogens is the key strategy for development of anti-infective agents. New findings from the complete human genome sequence as well as genomes of some pathogens and on-going transcriptome and proteome projects offer novel targets for the fight against infectious diseases. Therefore, it is necessary to accelerate the discovery of anti-infective agents by biochemical methodologies and to develop novel biologically active compounds.

Although DNA and RNA are known as the most important biological macromolecules responsible for encoding and transmitting genetic information, they have a wide range of applications to genetic testing, molecular biology and medicine. Oligonucleotides not only can serve as probes for detecting complementary DNA or RNA targets, but they can also act as an affinity probes or molecular recognition elements for a variety of targets including small molecules, sugar moieties, lipids, peptides, proteins and even whole cells.

Nucleic-acid-based scaffolds that can bind to various targets are called aptamers (including single stranded-DNA [ssDNA] or RNA). Oligonucleotide aptamers fold into three-dimensional shapes and bind noncovalently to a variety of biological targets with high affinity.1 Aptamers are functionally exploited as antagonists, agonists, or targeting ligands and are also called “chemical antibodies”. An anti-VEGF RNA aptamer (Macugen), the first aptamer therapeutic agent, was approved by the United Sates Food and Drug Administration (FDA) in 2005. From then on, a number of novel aptamer-based diagnostics and therapeutics have been undergoing clinical evaluation.2 , 3

Aptamers and the systematic evolution of ligands by exponential enrichment (SELEX) process

Aptamers are generated through a process of directed molecular evolution known as SELEX (Fig. 1 ).4 , 5 In a conventional SELEX process, the initial oligonucleotide (ssDNA or RNA) pool has conserved primer-binding sites at each end and a random sequence (30–50 nt) in the middle to achieve a mixture of 1014–1015 unique nucleic acid molecules. These oligonucleotides will fold into different secondary and tertiary structures, and they are then mixed with targets to form aptamer-target complexes. The target-bound aptamers are separated from unbound oligonucleotides by membrane filtration, magnetic beads, affinity columns, or capillary electrophoresis. The bound aptamers are amplified by PCR (DNA aptamers) or RT-PCR (RNA aptamers), and this enriched pool of aptamers is subjected to the next round of selection. Finally, the enriched aptamer sequences are identified by cloning and sequencing of individual clones, or alternatively, in recent years, high-throughput sequencing methods and bioinformatics analysis. During the SELEX process, several counterselections (negative-target selections) are added to eliminate nonspecific oligonucleotides. After 8-20 rounds of selection, specific aptamers that can bind to the target with high affinity will be obtained.

Fig. 1.

Fig 1

Schematic diagram of the SELEX process. The initial ssDNA or RNA pool is incubated with targets to form aptamer-target complexes. The target-bound aptamers are separated from unbound oligonucleotides and then amplified by PCR (DNA aptamers) or RT-PCR (RNA aptamers). This enriched pool of aptamers is used in the next round of selection. After the final round of selection, the enriched aptamer sequences are identified by cloning and sequencing individual clones.

Optimization of SELEX methods

To identify nucleic acid-protein interactions, genomic SELEX has been developed. Unlike randomized-sequence nucleic acids in conventional SELEX process, the starting libraries in genomic SELEX are derived from the genome of the organism of interest. Whole-cell SELEX is a method developed and modified for creating aptamers binding to live cells including bacteria. Because the targets in whole-cell SELEX are live tumor cells or pathogenic bacteria, aptamers can find and more effectively bind to the surface molecules of target cells (than those in conventional SELEX approaches) and should have little cross-reactivity with the nontargets.

Aptamer vs. antibody

Although aptamers are similar to antibodies in terms of their affinity and specificity to targets, they offer several advantages over their antibody counterparts. First, aptamers are selected through an in vitro process and do not depend on animals, cells, or even in vivo conditions. Antibody generation requires the use of a live animal to stimulate an immune response, and the target molecule needs to be immunogenic as well as nontoxic. Second, ssDNA aptamers are stable at room temperature, whereas antibodies require refrigeration to avoid denaturation. Third, aptamers have shown low immunogenicity and toxicity thus far,1 , 6 whereas antibodies may evoke a negative immune response. Finally, aptamers are small molecules and may effectively penetrate into tissue barriers and have effects on cells.

Aptamers are excellent non-protein-based alternatives to antibodies. In recent years, aptamers and the SELEX technology have received much more attention and been increasingly applied in the biomedical field, especially to the diagnosis and treatment of cancers and infectious diseases. In this review, we highlight areas where aptamers have been designed as diagnostic and therapeutic agents for major human infectious diseases.

Aptamers in diagnostics and therapeutics for bacterial infections

Aptamers targeting bacteria can be classified into the following two groups: (a) targeting predefined bacteria cell surface antigens or bacterial virulence factors; and (b) targeting whole cells with known or unknown molecular targets. To date, studies on antibacterial aptamers have been mostly focused on Mycobacterium tuberculosis (M. tb), Salmonella, Listeria monocytogenes (L. monocytogenes), Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) (Tables 1 and 2 ).

Table 1.

Summary of potential antibacterial aptamers for diagnosis.

Aptamer Type Organism Target Binding affinity Detection limit Ref
T9 DNA M.tb Beijing strains ManLAM Kd 7
668 ± 59 nM
CE24 DNA M. tb H37Rv CE protein Kd 8
CE15 CE24: 0.375 μM
CE15: 0.16 μM
CSIR 2.11 DNA M. tb CE protein 9
MPT64-A1 DNA M. tb MPT64 10,11
–(electrochemical aptasensor) DNA
M. tb MPT64 0.5 ± 0.2 fg/mL 12
–(electrochemical aptasensor) DNA M. tb MPT64 0.2 fg/mL 13
G43 RNA M. tb EsxG protein Kd 14
G78 G43: 8.04 ± 1.90 nM
G78: 78.85 ± 9.40 nM
MA1 DNA M. tb Whole-bacterium Kd 16
12.02 nM).
Aptamer 1 DNA M. tb Whole-bacterium Kd 100 CFU/mL 17
37 ± 4 nM
Aptamers 33 and 45 DNA S.Typhimurium Outer membrane proteins 3 CFU/ mL (Aptamers 33) 25,26
I-2 RNA S. Typhimurium OmpC protein Kd 28
20 nM
C4 DNA S. Typhimurium Whole-bacterium 31
ST2P DNA S. Typhimurium Whole-bacterium Kd 25 CFU/mL 32
6.33 ± 0.58 nM
SAL 26 DNA S. Typhimurium Whole-bacterium Kd 102 CFU/mL 33
123 ± 23 nM
Apt22 DNA Salmonella Paratyphi A Whole-bacterium Kd 103 CFU/mL 34
47 ± 3 nM
S25 RNA S. Enteritidis Mixtures of ten strains of S. Enteritidis 38
Crn-1 and crn-2 DNA S. Enteritidis Whole-bacterium Kd 35
crn-1:
0.971 µM
crn-2:
0.309 µM
APTSEB1 DNA S. aureus SEB 42
C10 DNA S. aureus SEC1 Kd 6 ng/mL SEC1 in food samples 43
65.14 ± 11.64 nM/L
R12.06 DNA S. aureus Alpha toxin Kd 200 nM alpha toxin in human serum samples 44
93.7 ± 7.0 nM
Antibac1 and Antibac2 DNA S. aureus Peptidoglycan Kd 45
Antibac1:
0.415 ± 0.047 μM
Antibac2:
1.261 ± 0.280 μM
PA#2/8 DNA S. aureus Protein A Kd 46
172 ± 14 nM for the recombinant Protein A
84 ± 5 nM for the native Protein A
SA20, SA23, SA31, SA34 SA43 DNA S. aureus Whole-bacterium Median effective concentration, EC50 682 CFU/mL in whole blood (SA20-conjugate-based assay) 47,48
SA20:
70.86 ± 39.22 nM
SA23:
61.50 ± 22.43 nM
SA31:
82.86 ± 33.20 nM
SA34:
72.42 ± 35.23 nM
SA43:
210.70 ± 135.91 nM
SA17 DNA S. aureus Whole-bacterium Kd Single cell 49
SA61 SA17: 35 nM
SA61: 129 nM
A8 DNA L. monocytogenes Internalin A 103 CFU/mL 55
LLO-3 DNA L. monocytogenes Listeriolysin O 4–61 CFU/100 μL 56
Lbi-17 DNA L. monocytogenes Whole-bacterium 60 CFU/500 μL 57
LMCA2 LMCA26 DNA L. monocytogenes Whole-bacterium Kd 20 CFU/mL 59
LMCA2:
2.01 × 10−12 M
LMCA26:
1.56 × 10−10 M
E-5, E-11, E-12, E-16, E-17, E-18, E-19 DNA E. coli O157 LPS 60
AM-6 DNA E. coli O157 Whole-bacterium Kd 63
107.6 ± 67.8 pmol
Apt B12 DNA E coli K88 Whole-bacterium Kd 1.1 × 103 CFU/mL in pure culture; 2.2 × 103 CFU/g in artificially contaminated fecal sample 64
15 ± 4 nM
E1, E2, E10, E12 DNA E. coli KCTC 2571 Whole-bacterium Kd 8 CFU/mL 65,68
E1: 12.4 nM
E2: 25.2 nM (E1)
E10: 14.2 nM
E12: 16.8 Nm
Ec3(31) RNA E. coli DH5α Whole-cell Kd 2 × 104 CFU/mL 69
225 nM
8.28A DNA E. coli DH5α Whole-cell Kd 70
27.4 ± 18.7 nM.

Table 2.

Summary of potential antibacterial aptamers for therapy.

Aptamer Type Organism Target Binding Affinity Detection limit Ref

NK2 DNA M. tb H37Rv Whole-bacterium K1a 18
1.84 × 105(± 1.5 × 104) M−1;
K2a
7.65 × 106 (± 6.0 × 105)  M−1
ZXL1 DNA M. tb H37Rv ManLAM Kd 19
436.3 ± 37.84 nM
BM2 DNA BCG ManLAM Kd 20
8.59 ± 1.23 nM
Mtb-Apt1 Mtb-Apt6 DNA M. tb Acetohydroxyacid synthase Kd 22
Mtb-Apt1:
1.06 ± 0.10 μM
Mtb-Apt6:
0.210 ± 0.05  μM
S-PS8.4 RNA S. Typhi Type IVB pili Kd 1 CFU 39,40
8.56 nM
S3 DNA S. aureus SEA Kd 51
36.93 ± 7.29 nM
AT-33 DNA S. aureus α-toxin Aptamers increased viability from 50–60% in cells treated with toxin alone up to about 85–90%. 52
AT-36

M. tb

Tuberculosis (TB) is the leading cause of deaths due to infectious diseases worldwide. Traditional diagnostic methods for TB, including tuberculin skin test, tuberculosis antibody detection and microscopic detection of M. tb, have some drawbacks, such as low sensitivity, poor specificity, lengthy procedure, and false-positive or -negative results. Bacterial culture, the “gold standard” of clinical tuberculosis diagnosis, required more than 10 days. Therefore, there is a clear need for new agents and strategies for TB diagnosis.

Aptamers targeting M.tb surface lipoglycan, proteins and whole bacterial cells have been generated as potential diagnostic tools. We selected ssDNA aptamer T9, which binds to mannose-capped lipoarabinomannan (ManLAM) from the predominant clinical epidemic M.tb strains of the Beijing genotype.7 ManLAM is the major surface lipoglycan of M. tb and has immunomodulatory activity. Aptamer T9 detects ManLAM antigens in serum and sputum samples from patients with active pulmonary TB (aPTB) and patients with extrapulmonary TB (EPTB).7 The 6 kDa early secreted antigenic target (ESAT6) and 10 kDa culture filtrate protein (CFP10) are secreted early by virulent M. tb and are not present in nonvirulent BCG. Both our group and Rotherham et al. have reported ssDNA aptamers against ESAT6 and CFP10 (CE protein).8 , 9 These aptamers can detect ESAT6 and CFP10 antigens in serum samples from patients with aPTB or EPTB8 or in sputum samples.9 MPT64, which is a 24 kDa protein secreted by M. tb during bacterial growth, is also used as a diagnostic target for aptamers. Qin et al. selected ssDNA aptamers against MPT64 protein from M. tb.10 They developed ssDNA aptamer MPT64-A1 against MPT64 antibodies, and reported that aptamer MPT64-A1 has the potential for the serological diagnosis of pulmonary TB, both in sputum smear-positive and -negative patients.11 Then several ssDNA aptamer-based electrochemical biosensors were devised for the detection of MPT64 and show the limit of detection in the fg mL−1range.12 , 13 Ngubane et al. demonstrated the suitability of surface plasmon resonance (SPR)-based SELEX for identification of two RNA aptamers, G43 and G78, which bind to the M. tb EsxG protein with high affinities.14 The EsxG protein is both encoded within the ESX-3 locus of M. tb and secreted by the ESX-3 system, which is essential for the optimal growth of pathogenic Mycobacteria in low-iron environments.14 They expected that these RNA aptamers would be candidate agents for the diagnosis of TB.14 Additionally, Russell et al. generated slow off-rate modified aptamers (SOMAmers) with subnanomolar affinity for 18 M. tb protein targets and evaluated their utility for direct antigen detection.15

Two groups have selected ssDNA aptamers against whole cells for M. tb H37Rv detection.16 , 17 Aimaiti et al. obtained species-specific aptamer MA1 against M. tb H37Rv and employed it in a sandwich enzyme-linked immunosorbent assay (ELISA).16 Zhang et al. selected an ssDNA aptamer against whole cells of M. tb H37Rv and immobilized the aptamer on an Au interdigital electrode (Au-IDE) of a multichannel series piezoelectric quartz crystal (MSPQC), then constructed a single-walled carbon nanotubes (SWCNT)/aptamer/Au-IDE MSPQC sensor for M. tb H37Rv detection17.

Several groups have conducted studies on aptamers for potential therapeutic applications against TB. Our group has selected several aptamers specifically binding to M. tb whole cells18 and surface lipoglycan.19, 20, 21 We first implemented whole-cell SELEX and generated ssDNA aptamer NK2, which binds to virulent M. tb H37Rv with high affinity and specificity.18 We then selected ssDNA aptamer ZXL1, which specifically binds to ManLAM from virulent M. tb strain H37Rv,19 , 21 and significantly reduces the progression of M. tb H37Rv infections as well as bacterial loads in mice and rhesus monkeys.19 Based on the different structures of ManLAM among different Mycobacteria, we next selected ssDNA aptamer BM2 that specifically binds to ManLAM from BCG.20 Aptamer BM2 acts as an adjuvant enhancing immunoprotective effects of BCG against virulent M. tb infection in murine and monkey models.20 To our knowledge, this was the first report regarding an aptamer serving as an adjuvant. Other groups have also reported aptamers potential for TB therapy. Baig et al. identified ssDNA aptamers Mtb-Apt1 and Mtb-Apt6 as a novel class of potent inhibitors of M. tb acetohydroxyacid synthase, which has been implicated in bacterial survival.22 Ranjbar et al. designed and constructed a three-dimensional DNA origami box based on MPT64 ssDNA aptamer for tuberculosis treatment.23 They expected that the MPT64 ssDNA aptamer-based drug delivery system would be capable of selectively interfacing with M.tb and would release an anti-TB drug near the bacteria.23

Salmonella

Salmonella infection arising from contaminated food has continued to be a concern for a decade because millions of cases of Salmonella infection occur worldwide annually. The detection of Salmonella before contaminated foods can be consumed is therefore an essential preventive measure for a public health system.24 Joshi et al. chose outer-membrane proteins of Salmonella Typhimurium (S. Typhimurium) as the selection target and generated aptamers 33 and 45.25 Aptamer 33 was conjugated to magnetic beads and used to capture S. Typhimurium seeded in whole-carcass chicken rinse samples, followed by detection via quantitative RT-PCR. They reported that 10–100 colony-forming units (CFUs) were successfully detected in 9 mL of the chicken rinsate in a pull-down assay.25 Then, aptamer 33 was tested by Ma et al. to generate an electrochemical biosensor for Salmonella detection, and the detection limit turned out to be as low as 3 CFU/mL.26 Aptamer 45 was chosen by Bagheryan et al. to develop a label-free impedimetric aptamer-based biosensor for S. Typhimurium detection.27 Han et al. identified an RNase-resistant RNA aptamer I-2 specifically targeting outer-membrane protein C (OmpC) of S. Typhimurium. This selected aptamer specifically binds to S. Typhimurium but not to gram-positive bacteria (S. aureus) or to other gram-negative bacteria (E. coli O157:H7).28

Several groups have used whole-cell SELEX to identify ssDNA aptamers binding to S. Typhimurium, Salmonella Paratyphi and Salmonella enterica serovar Enteritidis (S. Enteritidis) for Salmonella detection.29, 30, 31, 32, 33, 34, 35 These aptamers binding to the whole cell of Salmonella were conjugated to nanomaterials such as gold nanoparticles (AuNPs) and fluorescent nanoparticles as probes, magnetic nanoparticles (MNPs) as concentration elements, or loaded onto SWCNTs and reduced graphene oxide (rGO).29 , 30 , 34 , 36 , 37 Oh et al. immobilized AuNPs-ssDNA aptamer conjugates onto a localized surface plasmon resonance (LSPR)-based sensor chip and reported a method for detection of S. Typhimurium in spiked pork meat samples with the detection limit of 1.0 × 104 CFU/mL.30 Hyeon et al. tested mixtures of 10 strains of S. Enteritidis as targets and obtained RNA aptamer S25, which specifically binds to S. Enteritidis without any cross-reactivity with other Salmonella serovars.38

As for potential aptamers for treatment of Salmonella infection, we developed an effective RNA aptamer called S-PS8.4 for type IVB pili of Salmonella enterica serovar Typhi (S. Typhi).39 RNA aptamer S-PS8.4 inhibits the entry of a piliated S. Typhi strain into THP-1 cells, indicating that it has possible applications as a therapeutic agent.39 Based on S-PS8.4, Zelada-Guillén et al. reported a potentiometric biosensor for selective detection of a single CFU of S. Typhi, suggesting that the aptamer may also be applied to S. Typhi detection.40 Yeom et al. developed an aptamer-based drug delivery system for the treatment of S. Typhimurium infection.41 In this system, AuNPs are conjugated with an anti-S. Typhimurium ssDNA aptamer (AuNP-Apt), and antimicrobial peptides (AMPs) are loaded onto the AuNP-Apt. They reported antimicrobial activity of AMPs via AuNP-Apt conjugates during S. Typhimurium infection in vitro and in vivo.41

S. aureus

S. aureus is one of the major human pathogens that are responsible for a wide range of infections including hospital-acquired ones and life-threatening infections. In addition, the bacteria cause food-borne illnesses due to the excretion of several enterotoxins. Therefore, several ssDNA aptamers have been developed against staphylococcal enterotoxins (SEs). To develop potential diagnostic agents, aptamer APTSEB1 for staphylococcal enterotoxin B (SEB),42 aptamer C10 for S. aureus enterotoxin C1 (SEC1),43 aptamer R12.06 for α-toxin,44 aptamer Antibac1 and Antibac2 for a peptidoglycan45 and aptamer PA#2/8 for protein A46 were generated for the detection of SEs. Among these aptamers, aptamer R12.06 for α-toxin was utilized in a modified sandwich ELISA and sensitive detection of 200 nM a-toxin in undiluted human serum samples was achieved.44

The first ssDNA aptamers specific to whole cells for S. aureus detection were introduced by Cao et al. in 2009.47 They obtained a panel of ssDNA aptamers specific to S. aureus and have demonstrated that five aptamers (SA20, SA23, SA31, SA34 and SA43) recognize different molecular targets on S. aureus and have strong affinity, with K d values in the nanomolar range.47 SA20 was then chosen by Borsa et al. to develop a silica nanoparticle-aptamer conjugate, and they reported that their SA20-conjugate-based assay was successfully validated as a sensing platform for the specific detection of S. aureus cells at concentrations as low as 682 CFUs in 1 mL of whole blood.48 Chang et al. also generated ssDNA aptamers SA17 and SA61 specifically for S. aureus with nanomolar affinity.49 Aptamers SA61 and SA17 were conjugated with AuNPs and magnetic beads, respectively, and used in a resonance light scattering (RLS) spectral assay.49 They reported that the limit of detection of the assay is as low as a single S. aureus cell in tested samples within 1.5 h.49 Then, SA17 was employed by Wang et al. to develop a nanosystem for early sepsis diagnosis.50 Their nanosystem is capable of simultaneous blood bacterial species identification and enrichment in a single step, and they reported successful diagnosis of S. aureus infection in mice by means of this nanosystem.50

Some studies have been conducted to select aptamers for the treatment of S. aureus infection.51, 52, 53, 54 Aptamer S3, which binds staphylococcal enterotoxin A (SEA) with K d of 36.93 ± 7.29 nM, effectively inhibits SEA-mediated proliferation of human peripheral blood mononuclear cells and inflammatory secretion.[51] This result indicate that aptamer S3 is a potential agent for the treatment of S. aureus infection and SEA-induced diseases. α-Toxin is a major cause of S. aureus toxicity. SsDNA aptamers AT-33 and AT-36 against α-toxin were generated by Vivekananda et al. 52 They reported that these aptamers significantly inhibit α-toxin-induced cell death and cytokine upregulation in Jurkat T cells.52 Two research groups have employed anti-S. aureus ssDNA aptamers as the recognition elements in a drug delivery system.53 , 54 Kavruk et al. designed aptamer-gated nanocapsules for the specific targeting of S. aureus with a controlled release of antibiotics.53 Ocsoy et al. described the development of ssDNA aptamer-functionalized gold nanorods (Apt@Au NRs) for inactivation of methicillin-resistant S. aureus (MRSA) by targeted photothermal therapy (PTT).54 They immobilized the aptamers onto Au NRs and demonstrated that Apt@Au NRs act as a targeting and photothermal agent that can selectively recognize and effiectively inactivate MRSA.54

L. monocytogenes

L. monocytogenes is a foodborne gram-positive bacterium that causes Listeriosis: a serious illness that can even cause death. These bacteria can survive and grow in a wide range of environmental conditions, including refrigeration temperatures (4 °C). The United Sates FDA and the European Union have all implemented zero-tolerance policies toward L. monocytogenes in ready-to-eat foods, thereby leading to a substantial effort to develop highly sensitive biosensors for foodborne pathogen detection. Thus far, all anti-L. monocytogenes aptamers in recent studies were developed for bacterial detection. Ohk et al. chose internalin A (an invasin protein of L. monocytogenes) as a target molecule to generate ssDNA Aptamer A8. Alexa Fluor 647-conjugated A8 was used in a fiber-optic sensor together with an antibody in a sandwich format for the detection of L. monocytogenes in food.55 They demonstrated that A8 detects 103 CFU/mL L. monocytogenes in pure culture and in a mixture with other bacteria as well as L. monocytogenes from artificially contaminated (initial inoculation of 102 CFUs per 25 g) ready-to-eat meat products after 18 h of enrichment.55 Listeriolysin O (LLO) is a cytolysin expressed in L. monocytogenes. Bruno et al. developed an ssDNA aptamer called LLO-3 for the LLO protein.56 The aptamer was then applied to a fluorescent enzyme-linked DNA aptamer-magnetic bead sandwich assay for the detection of Listeria.56 They reported that the assay was highly sensitive, with limits of detection in the range of 4 to 61 L. monocytogenes cells or the equivalent LLO produced by 4–61 cells on average per 100 μL sample in separate titration trials.56

Some studies were conducted for the generation of ssDNA aptamers for whole cells of L. monocytogenes.57, 58, 59 Suh et al. reported that ssDNA aptamers for L. monocytogenes were identified after six rounds of positive and two rounds of negative selection.57 These ssDNA aptamers have binding affinity for L. monocytogenes with a micromolar K d value and only weak binding affinity for non-Listeria species.57 One aptamer, Lbi-17, was chosen for conjugation to magnetic beads and was employed in a combined aptamer magnetic capture (AMC)-qPCR assay. They demonstrated that the pathogen could be detected at concentrations < 60 CFUs per 500 μL of a buffer, with capture efficacy of 26–77%.57 One year later, Suh et al. demonstrated that they developed new ssDNA aptamers for L. monocytogenes.58 The affinitiy of these new ssDNA aptamers was improved, with K d values in the nanomolar range, and these aptamers were specific for L. monocytogenes in different growth phases.58 Lee et al. adopted a whole-cell SELEX strategy, generated ssDNA aptamers LMCA2 and LMCA26, and then developed an aptamer-based sandwich assay platform for the quantification of L. monocytogenes.59 They demonstrated that this platform yields a linear response over a wide concentration range of L. monocytogenes from 20 to 2 × 106 CFU/mL, thereby facilitating reliable detection of these bacteria at extremely low concentrations.59

E. coli

A lot of virulent strains of E. coli have been chosen as targets for the selection of specific aptamers because these bacteria are found ubiquitously in the intestinal flora of animals and humans, and their pathogenic variants cause major public health problems. E. coli O157:H7 causes several severe illnesses in human beings such as a gastrointestinal disease and bloody diarrhea (that is a root cause of hemolytic uremic syndrome). Bruno et al. generated ssDNA aptamers against E. coli O157 lipopolysaccharide (LPS),60 and these aptamers were then used to develop biosensors with chemiluminescence61 and color62 for rapid detection. Amraee et al. selected ssDNA aptamer AM-6 with higher sensitivity and specificity for recognition of whole-cell E. coli O157:H7 and reported that the binding affinity of aptamer AM-6 for the target bacteria is 107.6 ± 67.8 pM.63

An ssDNA aptamer library specific for E. coli K88 whole cells was enriched by Peng et al. for E. coli detection. They also developed a sandwich detection system, in which K88 monoclonal antibody-conjugated magnetic beads capture the target bacteria, and the ssDNA aptamers serve as the reporter in a fluorescence assay. They reported that this aptamer-based system detects bacteria at 1.1 × 103 to 2.2 × 103 CFU/g in samples.64 Kim et al. isolated and characterized ssDNA aptamers against E. coli KCTC 2571.65 During the selection, they used other bacterial species (Klebsiella pneumoniae, Citrobacter freundii, Enterobacter aerogenes, and Staphylococcus epidermidis) for counter-selection to enhance the selectivity of ssDNA aptamers for target E. coli detection.65 One of these aptamers (E2) was conjugated with fluorescent nanoparticles (A-FNPs) and was incorporated into an optofluidic particle-sensor platform for high-throughput real-time and single-cell detection of E.coli.66 Aptamer E2 was then employed by Jin et al. to develop a detection platform based on fluorescence resonance energy transfer (FRET) for rapid, ultrasensitive and specific detection of bacteria.67 Another aptamer against E. coli KCTC 2571, called aptamer E1, was applied to label-free and highly sensitive electro chemical detection of E. coli on the basis of rolling circle amplifications coupled peroxidase-mimicking DNAzyme amplification, with the detection limit of 8 CFU/mL.68 Dua et al. generated a 2‘F modified RNA aptamer Ec3 by whole-cell SELEX to target E. coli DH5α.69 They developed an aptamer Ec3-based gap capacitance biosensor for electrochemical impedance spectroscopy (EIS); it has the detection limit of 2 × 104 CFU/mL for E. coli cells without any labeling and signal amplification techniques.69 Renders et al. conducted whole-cell SELEX to select ssDNA aptamers with a tyrosinelike side chain against live E. coli DH5α cells.70 They reported that aptamer 8.28A possesses high selectivity and affinity for the target cells and was greatly enriched for phenolmodified dU nucleotides.70 This result highlights a means of increasing the chemical diversity of aptamers that will afford high affinity and specificity to potentially challenging targets.70

The reports on screening of aptamers as potential antibacterial or “antibiotic” agents against infection by virulent E. coli are scarce. DNA aptamers were developed against LPS from E. coli O111:B4 by Bruno et al. 71 They demonstrated that their polyclonal anti-LPS aptamers bind both LPS and E. coli with high affinity, and these aptamers were coupled to the human C1qrs protein and triggered destruction of E. coli O111:B4 and K12 strains.71

Aptamers in the diagnostics and therapeutics for viral infections

Specific aptamers can target the viral proteins involved in the stages of adsorption, penetration, replication, maturation and even release during a viral infection cycle. Currently, some aptamers have been are reported to have potential applications in the diagnostics and therapeutics for viral infections, e.g., human immunodeficiency virus-1 (HIV-1), hepatitis C virus (HCV), influenza virus (Tables 3 and 4 ).

Table 3.

Summary of potential antiviral aptamers for diagnosis.

Aptamers Type of aptamer Organism Target Binding Affinity (Kd) Inhibitory effect Ref
RNATat RNA HIV-1 Tat 120 ± 13 pM Strong affinity for Tat even in the presence of a large excess of HIV TAR in cell culture 73, 74, 75
ZE2 DNA HCV E2 protein 1.05 ± 1 nM Inhibition of HCV cell culture (HCVcc) infection in vitro 108
9-14 RNA HCV Core protein 9–14: 142 nM; 109
9-15 9–15: 224 nM
C4 DNA HCV Core Protein Inhibition of HCV production 110,111
LD: 3.3 pg/mL
A22 DNA Influenza A virus (H3N2) HA(91-261) Inhibition of the hemagglutinin capacity of the virus and viral infectivity in vitro 128
Clone B RNA Influenza A virus (H3N2) HA 200 pM 129
P30-10-16 RNA Influenza A virus (H3N2) Whole virus 188 pM 95% inhibition of viral fusion efficiencies in the presence of 5 μM aptamers 130
A10 DNA Influenza A virus (H5N1) HA Inhibition of receptor binding 131
C7 DNA Influenza A virus (H9N2) HA(101-257) 55% inhibition of the viral infection at 1 nM in the cell viability assay 132
HAS15-5 RNA Influenza A virus (H5N1) HA Inhibition of receptor binding 133
D-26 RNA Influenza A virus (H1N1) HA 67 fM Complete inhibition of the agglutination of RBC in the presence of 200 nM aptamer 134
HA68 DNA Influenza A virus (H3N2) HA 7.1 nM Inhibition of the agglutination of RBC in the presence of 2.5 μM aptamers, and detection of H3N2 variant isolated from human cases 135
8-3 RNA Influenza A virus (H5N1 and H7N7) HAs from H5N1 and H7N7 170 fM Efficiently interferes with HA-glycan binding (EC50: 25 nM) 137
ApI DNA Influenza A virus HA1 subunit ApI: 64.76 ± 18.24 nM Binding to the HA1 subunit of subtype H1 (H1-HA1), but not to the HA1 subunit of subtype H5 (H5-HA1) 138
ApII ApII: 69.06 ± 12.34 nM
ApIII ApIII: 50.32 ± 14.07 nM
A9 DNA Influenza A virus (H9N2) HA A9: 46.23 ± 5.46 nM Inhibition of H9N2 virus infection in vitro 139
B4 B4: 7.38 ± 1.09 nM
DNA Influenza A virus Whole virus Kd 142
(H1N1) 55.14 ± 22.40 nM
IF22 DNA Influenza A virus (H5N1 and H5N8) Whole virus 144
IF23
HBs-A22 RNA HBV Surface antigen Inhibition of receptor binding 147
G5α3N.4 RNA HPV 16 E7 protein 1.9 μM 152

Table 4.

Summary of potential antiviral aptamers for therapy.

Aptamers Type of aptamer Organism Target Binding affinity (Kd) Inhibitory effect Ref
B40 RNA HIV-1 Gp120 B40: 21 ± 2 nM Inhibition of gp120-CCR5 interaction 78
B40t77 B40t77: 31 ± 2 nM
A-1 RNA HIV-1 Gp120 52 nM Inhibition of HIV-1 infection in primary human peripheral blood mononuclear cells 80
BclON-mut DNA HIV-1 Gp120 143 ± 79 nM IC50: 86
BclON-mut 0.08 ± 0.03 μg/mL
F-thio-BclON 86 ± 17 nM F-thio-BclON 0.040 ± 0.003 μg/mL
1.1 RNA HIV-1 RT 5 nM 88
RT1t49 DNA HIV-1 RT 1 nM IC50: 0.3 nM 89
4.20 DNA HIV-1 RT 180 ± 70 pM 90
R12-2 DNA HIV-1 RT 70 nM Inhibition of the RNase H activity of intact HIV-1 RT; IC50 < 100 nM 91
37NT DNA HIV-1 RT 660 pM Inhibition of primer-template binding 93
FA1 FANA aptamer HIV-1 RT Low pM range Inhibition of HIV-1 RT primer extension 95
RNApt16 RNA HIV-1 5′-untranslated region of HIV-1 genome 280 ± 60 nM Inhibition efficiency in a human cell line: 85% 96
IV04 DNA HIV-1 TAR RNA element 20 nM Formation of RNA-DNA kissing complexes for disrupting TAR secondary structure 97
T30695 DNA HIV-1 Integrase 0.5 ± 0.2 μM IC50 < 100 nM 98,102
93del ssDNA HIV-1 Integrase Inhibition of HIV-1 integrase in the nanomolar range 99, 100, 101
8-6 RNA HIV-1 Nucleo-capsid protein 1.4 × 10−9 M 103
DP6-22 RNA HIV-1 Gag protein DP6-22: 100 ± 3.4 nM Disrupting Gag-genomic RNA interaction and negatively affectiong genomic RNA transcription, processing, or stability 104
RBE(apt) RNA HIV-1 Rev protein Conjugation with ribozyme targeting HIV Env for gene therapy 105
S3R3 RNA HIV-1 Integrase 47 ± 3 nM Anti-integrase aptamer expressed as an shRNA–aptamer fusion confered long term resistance to HIV-1 replication in T cells. 106
G6-16 RNA HCV NS3 protein 238 nM IC50: 3 μM 112
G9-I RNA HCV Truncated protease domain of NS3 protein 10 nM IC50: 100 nM 113
G5 RNA HCV Helicase domain of NS3 25 nM IC50: 50 nM 116
3-07 RNA HCV IRES domains III–IV 9 nM Inhibition of IRES-dependent translation 117
AP50 RNA HCV IRES 5 nM Inhibition of IRES-dependent translation; IC50 value in the low μM range 118, 119
HH-11 RNA HCV IRES domain IIIf and IV IC50 value of 170 ± 20 nM 120
27v DNA HCV NS5B 132.3 ± 20 nM IC50: 196 ± 16 nM 121,122
r10/43 RNA HCV NS5B r10/43: 1.3 ± 0.3 nM Ki 123
r10/43: 1.4 ± 2.4 nM
r10/47 r10/47: 23.5 ± 6.7 nM r10/47: 6.0 ± 2.3 nM
R-F t2 RNA HCV NS5B 2.62 ± 0.90 × 10−9 M Blocking both the initiation and the elongation of RNA synthesis 124,125
HA12-16 RNA Influenza A virus (H5N2) Glycosylated HA Preventing influenza infection by strongly binding to the glycosylated HA 145
PAN-2 DNA Influenza A virus Amino acid residues in the N-terminal of the PAN of the influenza A virus polymerase Kd IC50: around 10 nM 146
(H1N1, H5N1, H7N7, and H7N9) 247 ± 11 nM
S9 RNA HBV Truncated P protein Competitively inhibiting P protein binding and suppressing viral pgRNA packaging 148
Apt.No.28 DNA HBV Core protein Inhibiting the assembly of the nucleocapsid; reducing extracellular HBV DNA 149
AO-01 DNA HBV Capsid 180 ± 82 nM 47% inhibition of virion production at 3 days post transfection in vitro 150
A2 RNA HPV 16 E7 protein 107 nM Inhibtion of E7-pRb interaction 151,153
F2 RNA HPV 16 E6 protein Inhibition of the interaction between E6 and PDZ-binding motif 154
NG8 DNA SARS-CoV Helicase 5 nM IC50: 91.0 nM 155
S15 DNA DENV-2 Envelop protein domain III 200 nM IC50: 4.2 μM 156
GE54 DNA RABV Glyco-protein 307 nm Inhibiting of viral replication and protecting mice challenged with RABV in vivo 157

HIV-1

HIV-1 is the cause of the majority of HIV infections globally. HIV infection leads to chronic viral disease and eventually to acquired immunodeficiency syndrome (AIDS). People with chronic HIV-1 infection may have none of the HIV-related symptoms for many years. Therefore, early detection of HIV-1 infection is important for HIV/AIDS treatment. HIV accessory proteins include the trans-activator of transcription (Tat) protein, which is important for the transcription of viral RNA and enhances the amount of protein produced by attaching itself to the viral RNA.72 The Tat protein is one of the most promising candidates for HIV screening because this protein is released in the body at an early stage of infection.72 Yamamoto et al. first generated an RNA aptamer specific for the HIV-1 Tat protein.73 The aptamer was then used to develop biosensors detecting the Tat protein.74 , 75 Another research conducted by Rahim Ruslinda et al. yielded a split RNA aptamer as the detection probe for Tat.76 They demonstrated that the effectiveness of a diamond-FET (field-effect transistor)-based RNA aptamer for the detection of a real sample of HIV-1 Tat protein is at concentrations down to 1 nM.76

Although combination antiretroviral therapy (cART) can suppress the HIV-1 virus and stop the progression of HIV-1 disease, it is not curative. Aptamers have been considered an alternative or adjuvant to the chemical antiviral agents in cART. Aptamers specifically targeting various parts of the HIV-1 genome and HIV-1 proteins have been selected and shown to effectively inhibit viral fusion with the target cell and to suppress viral replication. There are some aptamers available for HIV-1 envelope glycoprotein gp120. 77, 78, 79, 80 One of these RNA aptamers for gp120, called B40, blocks gp120 binding to its T cell co-receptor, CCR5 (C—C chemokine receptor type 5).78 , 81 Its shorter variant, B40t77 and modified B40t77 (UCLA1) have been shown to strongly neutralize HIV-1 infectivity.79 , 82 , 83 Another anti-gp120 aptamer, A-1, was applied to construct a therapeutic aptamer-siRNA chimera for HIV-1, and the chimera was reported to show therapeutic efficacy against of HIV-1 infection in vivo. 80 , 84 , 85 Recently, new potent anti-HIV-1 ssDNA aptamers that bind to gp120 were obtained by Prokofjeva et al. 86 They demonstrated that both G-quadruplex(G4)-forming aptamer (F-thio-BclON) and non-G4-forming aptamer (BclON-mut) inhibit lentiviral transduction in the low nanomolar range and thus can be considered for the development of microbicides against HIV-1.86 For inhibiting the HIV-1 replication cycle, reverse transcriptase (RT) is one of the main drug targets in HIV/AIDS. HIV-1 RT catalyzes the conversion of the single-stranded genomic viral RNA into double-stranded proviral DNA, which in turn is integrated into the host genome.87 To date, numerous RNA or DNA aptamers against HIV-1 RT have been generated, and their application to the inhibition of viral replication has been investigated.88, 89, 90, 91, 92, 93, 94, 95 Among these anti-RT aptamers, an ssDNA aptamer generated by capillary electrophoresis-SELEX and 2′-deoxy-2′-fluoroarabinonucleotide (FANA) aptamer FA1 can strongly bind HIV-1 RT, with low picomolar dissociation constants.90 , 95 Besides anti-RT aptamers, highly specific aptamers—that target various parts of the HIV-1 genome, the trans-activation-responsive (TAR) RNA element, or HIV-1 integrase, nucleocapsid, Gag or Rev proteins—have been isolated and shown to effectively suppress viral replication.96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106

HCV

HCV is the common cause of chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma.107 There is currently no approved HCV vaccine, and aptamer research for the diagnosis and treatment in this area is at the initial stages. For inhibiting HCV initial attachment to host cells as well as to develop agents for the early diagnosis of HCV infection, our group carried out cell surface-SELEX and generated ssDNA aptamer ZE2 that binds specifically to HCV surface glycoprotein E2.108 A good correlation was observed among HCV patients between the HCV E2 antigen-aptamer (ZE2) assay and the assays for HCV RNA quantities or HCV antibody detection.108 Additionally, aptamer ZE2 specifically captures HCV particles and significantly inhibits HCV cell culture (HCVcc) infection of human hepatocytes (Huh7.5.1) in vitro.108 We can speculate that ZE2 may be beneficial as a new molecular probe for HCV detection as well as anti-HCV therapy. Other research groups reported aptamers against HCV core (C) antigen for HCV detection.109, 110, 111 Ghanbari et al. immobilized an anti-C antigen aptamer onto graphene quantum dots (GOD), and the aptamer-GOD was placed on the surface of a glassy carbon electrode.111 They demonstrated that the electrochemical aptamer-based biosensor effectively detects the HCV C antigen with the detection limit of 3.3 pg/mL.111 These aptamers against HCV E2 and C antigens are crucial for the early diagnosis of hepatitis C within the “diagnostic window period,” when serum antibodies have not appeared yet.

In the earlier studies on aptamers for anti-HCV therapy, most efforts had been focused on developing novel aptamers binding to the HCV NS3 protein or the internal ribosome entry site (IRES).112, 113, 114, 115, 116, 117, 118, 119, 120 The NS3 protein possesses protease, nucleoside triphosphatase, and helicase activities. HCV IRES directs the binding of ribosomes in close proximity to the start codon of the viral open reading frame.112 , 117 The aptamers blocking the activities of NS3 or IRES are expected to inhibit HCV replication and translation and hold promise as anti-HCV therapeutic agents.112, 113, 114, 115 , 117 Recently, Romero-López et al. designed multivalent aptamers against the HCV genome.120 They reported that chimeric RNA aptamer called HH-11, with an IC50 value of 170 ± 20 nM, simultaneously targets HCV IRES domains IIIf and IV.120 The blocking of RNA-dependent RNA polymerase (nonstructural protein 5B [NS5B]) is another strategy to develop anti-HCV therapeutic aptamers. Aptamers against NS5B strongly inhibit HCV polymerase activity and viral replication. These aptamers suppress formation of infectious virus particle in vitro.121, 122, 123, 124, 125

Influenza virus

Influenza viruses cause respiratory diseases in birds and mammals. The three major types of influenza viruses are influenza A, B, and C. The influenza A virus leads to seasonal epidemics as well as sporadic pandemics of influenza, and hence it is regarded as the major cause of this acute infectious disease and deaths. In recent years, influenza rapid diagnostic tests became available. Most of these diagnostic tests are based on detection influenza antigens by antibodies.126 However, aptamers currently are viewed as equal or better tools compared with antibodies for antigen identification.127 The hemagglutinin (HA) antigen is ubiquitously expressed on the surface of influenza viruses and serves as the major target for aptamers. The HA antigen also facilitates the first stage of the influenza virus infection: the binding to a species-specific host cell in the respiratory epithelium. Thus far, more than 10 aptamers against HA have been developed for diagnostic purposes.128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139 RNA aptamer P30-10-16 binds to HA (human influenza A virus subtype H3N2) with more than 15-fold stronger affinity as compared with a conventional anti-HA monoclonal antibody.130 Bai et al. developed a portable SPR aptasensor (an aptamer-based biosensor) based on the specific DNA aptamer for the rapid detection of avian influenza virus (AIV) H5N1. The total detection time was < 1.5 h, which is faster than all conventional methods for AIV detection, such as virus isolation and identification (5–7 days), ELISA (3 h), and RT-PCT (3–5 h).140 Wang et al. selected an ssDNA aptamer against HA from AIV H5N1, and the K d value of this aptamer was 4.65 nM.136 The anti-HA aptamer was later immobilized onto a gold interdigitated microelectrode for the development of a rapid H5N1 detection method, and the method had the detection limit of 1 hemagglutination unit (HAU) in the tracheal chicken swab samples spiked with the H5N1 virus.141 Other aptamers for influenza virus detection target whole virus particles.142, 143, 144 Lai et al. carried out screening and found an influenza A virus-specific aptamer,142 which was later incorporated into an integrated microfluidic chip for H1N1 detection, and the limit of detection was found to be 0.032 HAU.143

As for potential therapeutic aptamers, Kwon et al. isolated an RNA aptamer specific to the glycosylated receptor-binding domain of the HA protein (gHA1) and demonstrated that the selected aptamer suppresses viral attachment to host cells by neutralizing the receptor-binding site of influenza virus HA.145 Yuan et al. chose amino acid residues in the N-terminus of the PA subunit (PAN) of the influenza A virus polymerase as a target to select ssDNA aptamers. They generated ssDNA PAN-2, which affords cross-protection against infections with H1N1, H5N1, H7N7, and H7N9 influenza viruses, with IC50 of ∼10 nM.146

Other viruses

Although it has been more than 20 years since aptamers were first constructed, the studies on the exploitation of aptamers against many viral diseases are not advanced enough and are not designed systematically. However, recently, a few studies were conducted on aptamers against some viruses such as hepatitis B virus (HBV),147, 148, 149, 150 human papilloma virus (HPV),151, 152, 153, 154 severe acute respiratory syndrome coronavirus (SARS-CoV),155 dengue viruses (DENVs)156 and rabies virus.157 , 158 Among these selected aptamers, several aptamers target viral surface antigens or virus-infected cells, e.g., RNA aptamer HBs-A22 specifically binds to HBV surface Ag-expressing hepatoma cells,147 ssDNA aptamer S15 targets DENV-2 envelope protein,156 and ssDNA aptamer GE54 targets neurotropic rabies virus (RABV) glycoprotein-expressing cells and rabies virus-infected cells.157 , 158 Some aptamers bind to viral enzymes to inhibit replication, e.g., an RNA aptamer against HBV polymerase protein148 and an ssDNA aptamer against SARS-CoV helicase.155 Several aptamers target viral proteins thus blocking nucleocapsid formation, e.g., ssDNA aptamers against the HBV core protein or matrix-binding domain.149 , 150 Other aptamers target viral oncoproteins, e.g., RNA aptamers binding to oncoproteins HPV16 E7 and E6.151, 152, 153, 154

Aptasensor methods

Aptasensors are biosensors in which the biological recognition elements are DNA or RNA aptamers.159 In recent years, more and more aptasensors have been designed for possible diagnostic applications. Major detection methods include (a) optical transduction, (b) electrochemical detection and (c) mass sensitive detection. In the following subsection, the basic principles of aptasensor-based detection are briefly discussed.

Optical transduction

Optical transduction can be subdivided into three major methods: absorbance assay, fluorescence detection and colorimetric detection. In an absorbance assay, an enzyme-linked oligonucleotide assay (ELONA) is usually implemented. In the direct method of ELONA, a plate is coated with the target molecule or cell, and a biotinylated aptamer binds to the target, followed by the addition of a streptavidin-horse radish peroxidase (HRP) conjugate and an enzyme substrate for color development (Fig. 2 A).7 , 8 , 19 In a sandwich assay of ELONA, the capture antibody or aptamer is immobilized on a plate, followed by addition of the target molecule, the HRP-linked aptamer or antibody, and an enzyme substrate. 7 , 8 , 20

Fig. 2.

Fig 2

Optical aptasensor. A: Direct ELONA. The target is coated onto the plate and the biotinylated aptamer binds to the target followed by the addition of streptavidin-HRP conjugate and enzyme substrate for color development. B: Fluorescent aptasensors. Left panel, aptamers are labeled with fluorophores and the changes in the fluorescent signal are detected upon targets bound by the aptamers. Right panel, FRET process. A quencher is labeled on the complementary sequence of the aptamer. When the aptamer binds to the target, the quencher is moved away from the fluorophore owing to the conformational change of the aptamer and affords “signal on” detection. C: Optical and AuNPs-based aptasensors. Aptamer1-AuNPs and Aptamer2-magnetic nanoparticles are mixed with the target bacteria. Aptamer2-magnetic nanoparticles-target-Aptamer1-AuNPs complexes are formed. In the magnetic field, the target bacteria are accumulated, and the changes in the absorbance spectrum are detected.

The principle of fluorescence detection is measurement of fluorescence signals (generation or quenching) when an aptasensor binds to a target (Fig. 2B). Aptamers can be labeled with fluorophores or conjugated with quantum dots (QDs), and the changes in the fluorescent signal will be detected after the target is bound by an aptamer.20 , 108 , 160 , 161 Sometimes, a FRET-based technology has been used in aptasensors. In a FRET process, fluorescent particles are donors and quencher particles are acceptors. The fluorescence of the donors can be quenched by the acceptors if they come close to each other (< 10 nM).162 A quencher is usually attached to the complementary sequence of the aptamer. Thus, the fluorescence of the aptamer can be quenched due to the good overlap between the fluorescence emission of the aptamer and the absorption spectrum of the complementary sequence. When an aptamer binds to the target, the quencher is moved away from the fluorophore owing to the conformational change of the aptamer and affords “signal on” detection.

AuNPs are widely used in aptasensor-based colorimetric assays. AuNPs have size-and-distance-dependent optical features. A well-dispersed AuNP solution is red, while the aggregated form of AuNPs is blue or purple due to the change in the surface resonance frequency of AuNPs.36 Aptamers are modified with a mercapto group at the 5′ or 3′ end and are conjugated with AuNPs through Au–S bonding. The binding of aptamer-AuNP conjugates to targets cause aggregation of the AuNPs, resulting in absorbance spectral changes of the AuNP solution (Fig. 2C).36 , 163 Sometimes, biotinylated aptamers are combined with streptavidin-coated AuNPs in reporter systems. In such a system, target bacteria are captured by a capture antibody or aptamer, and then, biotinylated aptamers and streptavidin-coated AuNPs are added develop color.164

Electrochemical detection

The principle of electrochemical detection is based on motoring the changes in electrical properties when an aptamer binds to a target. One strategy of the detection is to coordinate aptamers with SWCNTs for signal amplification (Fig. 3 A). SWCNTs serve as effective ion-to-electron transducers because of their extraordinary ability to promote electron transfer between heterogeneous phases of SWCNTs.40 , 165 , 166 In a SWCNT-based aptasensor system, aptamers are immobilized on SWCNTs. When the immobilized aptamers capture the target bacteria, a measurable change emerges in the conductance of the system because the SWCNT-aptamer hybrid material allows for a charge transfer between the SWCNT or a solution interface and the ions around the target bacterial wall.40 , 165 , 166

Fig. 3.

Fig 3

Electrochemical aptasensor. A: SWCNT-based aptasensor. Aptamers are immobilized on SWCNTs. When the immobilized aptamers capture the target bacteria, a measurable change emerges in the conductance of the system. B: GO-based aptasensor. Aptamers are loaded onto the GO sheet. The binding of the aptamers to targets causes the aptamers to leave from the sheet surface. Consequently, EMF changes take place.

Chemically modified graphene oxide (GO) and reduced graphene oxide (rGO) have excellent electrochemical and transduction properties and can be incorporated into an electrochemical aptasensor to enhance signals. Aptamers are covalently or noncovalently attached onto the GO/rGO sheet (Fig. 3B).167 The binding of the aptamers to targets causes the aptamers to leave the GO/rGO surface. Consequently, changes in the electromotive force (EMF) take place between the terminals of the GO/rGO aptasensor (as the working electrode) and the double junction reference electrode because of the separation of negatively-charged phosphodiester groups from the GO/rGO surface (Fig. 3B).167

Mass sensitive detection

Mass sensitive detection methods for pathogens include (a) an SPR aptasensor, (b) a surface acoustic wave (SAW) aptasensor and (c) a quartz crystal microbalance (QCM) aptasensor. These detection systems do not require additional labeling. In an SPR-based system, aptamers are usually labeled with biotin and immobilized on a streptavidin coated gold chip. The binding between targets and the aptamers leads to a change in the plasmon resonance and plasmon angle, which can be measured (Fig. 4 A).140 Both SAW aptasensors and QCM aptasensors are acoustic wave sensors. SAW aptasensors record the phase shift and amplitude change due to the binding of targets to surface-immobilized aptamers (Fig. 4B).168 In a QCM aptasensor system, aptamers are loaded onto gold-coated quartz. The binding of targets to aptamers causes an increase of the mass on the surface of the crystal and generates a detectable decrease in the resonance frequency of the crystal.169

Fig. 4.

Fig 4

SPR aptasensor and SAW aptasensor. A: SPR aptasensor. Aptamers are immobilized on the chip. The binding between targets and aptamers leads to a change in the plasmon resonance and plasmon angle. B: SAW aptasensor. The phase shift and amplitude of SAW changes because of the binding of targets to surface-immobilized aptamer.

Challenges of aptamer-based diagnostic and therapeutic applications

Although aptamers are promising candidates for diagnostic and therapeutic applications, some challenges still remain.

In vitro selection vs. in vivo targeting

Most aptamers have been selected in vitro. Given that the structures of some target molecules are diverse and complex in vivo, the aptamers may not work in vivo as expected. Cross-binding to non-target molecules may limit the use of aptamers for detection in many real world complex samples, such as biological fluids and food matrices. Therefore, careful design of the selection process is necessary to improve specificity of the identified aptamers, which can better distinguish closely related molecules at low concentrations. High variation of bacteria and viruses and complex structures of targets are additional factors that may influence the performance of aptamers. It is important to continue to develop simpler and more efficient SELEX procedures (requiring less time in selection) to develop specific and/or universal aptamers against various bacterial pathogens.

Nuclease degradation

Unmodified aptamers (especially RNA aptamers) are generally susceptible to digestion by cellular nucleases present in body fluids or cells. Therefore, chemical modifications are often introduced into the oligonucleotides to increase in vivo stability of aptamers. These chemical modifications include capping the 3-end with inverted thymidine/biotin,170, 171, 172, 173 phosphorothioate/methylphosphonate substitution in the phosphate backbone,174 2′-sugar modifications with a fluoro (F), amino (NH2) or O-methyl (OCH3) groups and locked nucleic acid (LNA) modification.175 The LNA modification is an intramolecular 2′-O to 4′-C methylene bridge, and LNAs show high resistance to ribozymes.175 The most common modifications are a substitution of a sulfur atom for a non-bridging oxygen in the DNA or RNA phosphate backbone as well as the modifications of RNA aptamers at the 2′-position on the ribose ring. Modified RNA aptamers have half-life ranging from several to hundred hours but unmodified RNA usually has a half-life of seconds in human serum.176

Renal filtration

Because of small mass (6–30 kDa) and short diameter (< 5 nm) of aptamers, they are easily filtered through the glomerular capillaries. To extend the circulation time of aptamers, they are conjugated to compounds with high molecular mass, e.g., polyethylene glycol (PEG),124 cholesterol,125 an antibody,177 liposome178 or other nanomaterials.179 PEG has been widely been used to prolong circulation half-life of biological agents. Besides half-life extension in the blood circulation, conjugation of PEG can improve drug solubility and stability.180 The first aptamer-based drug (Macugen) is based on the PEGylation.180 This PEGylated aptamer has a half-life of 9.3 h according to the pharmacokinetic experiments performed on rhesus monkeys.181

Conclusion

Over the last two decades, there has been a growing number of studies on aptamers targeting pathogens. Although few aptamers against fungi have been reported,182 , 183 a lot of aptamers that specifically target bacterial or viral virulence factors—as well as entire bacterial cells or viral particles—have been generated. For an example, ssDNA aptamer ZE2 against HCVsurface glycoprotein E2 has been used in several reports.184, 185, 186 Oligonucleotide aptamers are promising candidates for the diagnosis and treatment of infectious diseases.

The development of aptasensors is becoming an increasingly diverse field. In aptasensors, DNA or RNA aptamers serve as biological recognition elements, and an enzyme-linked oligonucleotide assay, fluorescence, colorimetry, electrochemistry and mass sensitivity have been popular as readout formats. The many attractive features of aptasensors encourage researchers to continue to optimize their use for disease diagnosis and treatment. An increasing number of applications of aptasensor to the realm of medicine may be commercialized in the future.

Aptamers have several advantages over protein antibodies: lower immunogenicity, easier production, and smaller size. Both aptamers and antibodies can potentially serve as diagnostic and therapeutic tools. Since the first monoclonal antibody was produced in the 1970s, more widespread distribution and adoption of antibodies as well as a well-developed infrastructure have led to widespread application of antibodies to disease diagnosis. However, aptamers are more stable and easier to label and modify, and have a much lower cost of manufacture as compared with antibodies. Therapeutic aptamers have the advantages over antibodies in the treatment of antitoxin, antivenom and anti-multiple drug resistant bacterial and viral infections, but the therapeutic effects and safety of aptamers should be carefully studied. Recently, the first report of aptamer-specific resistance in cell culture has been published, and the utility of broad-spectrum aptamers for medical treatment and tracking of long-term effects of aptamers may be necessary.187

The aptamer field has probably touched only the tip of the iceberg. Researchers are focus on optimization of the SELEX methods, aptamer modifications, combinations of aptamers and targeted drug delivery. The aptamer technology continues to reveal its promising feature and the vast diagnostic and therapeutic potential for infectious diseases in the present and the future.

Acknowledgments

Conflicts of interest

The authors declare that they have no conflict of interest.

Acknowledgments

This work was supported by Grants from the National Grand Program on Key Infectious Disease (2017ZX10201301-006), theNational Natural Science Foundation of China (81471910, 31770145, 31370197, 21572173, 81501377 and 91740120), the National Outstanding Youth Foundation of China (81025008), the Major Projects of Technological Innovation of Hubei Province (2016ACA150 and 2016CFA062), the Science and Technology Program of Wuhan (201150530141) and the Wuhan Youth Science and Technology Chen'guang Plan (2016070204010127).

References

  • 1.Sun H, Zu Y. A highlight of recent advances in aptamer technology and its application. Molecules. 2015;20:11959–11980. doi: 10.3390/molecules200711959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Sundaram P, Kurniawan H, Byrne ME, Wower J. Therapeutic RNA aptamers in clinical trials. Eur J Pharm Sci. 2013;48:259–271. doi: 10.1016/j.ejps.2012.10.014. [DOI] [PubMed] [Google Scholar]
  • 3.Bruno JG. Predicting the uncertain future of aptamer-based diagnostics and therapeutics. Molecules. 2015;20:6866–6887. doi: 10.3390/molecules20046866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990;346:818–822. doi: 10.1038/346818a0. [DOI] [PubMed] [Google Scholar]
  • 5.Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 1990;249:505–510. doi: 10.1126/science.2200121. [DOI] [PubMed] [Google Scholar]
  • 6.Ireson CR, Kelland LR. Discovery and development of anticancer aptamers. Mol Cancer Ther. 2006;5::2957–2962. doi: 10.1158/1535-7163.MCT-06-0172. [DOI] [PubMed] [Google Scholar]
  • 7.Tang XL, Wu SM, Xie Y, Song N, Guan Q, Yuan C. Generation and application of ssDNA aptamers against glycolipid antigen ManLAM of Mycobacterium tuberculosis for TB diagnosis. J Infect. 2016;72:573–586. doi: 10.1016/j.jinf.2016.01.014. [DOI] [PubMed] [Google Scholar]
  • 8.Tang XL, Zhou YX, Wu SM, Pan Q, Xia B, Zhang XL. CFP10 and ESAT6 aptamers as effective Mycobacterial antigen diagnostic reagents. J Infect. 2014;69:569–580. doi: 10.1016/j.jinf.2014.05.015. [DOI] [PubMed] [Google Scholar]
  • 9.Rotherham LS, Maserumule C, Dheda K, Theron J, Khati M. Selection and application of ssDNA aptamers to detect active TB from sputum samples. PLoS One. 2012;7:e46862. doi: 10.1371/journal.pone.0046862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Qin L, Zheng R, Ma Z, Feng Y, Liu Z, Yang H. The selection and application of ssDNA aptamers against MPT64 protein in Mycobacterium tuberculosis. Clin Chem Lab Med. 2009;47:405–411. doi: 10.1515/CCLM.2009.097. [DOI] [PubMed] [Google Scholar]
  • 11.Zhu C, Liu J, Ling Y, Yang H, Liu Z, Zheng R. Evaluation of the clinical value of ELISA based on MPT64 antibody aptamer for serological diagnosis of pulmonary tuberculosis. BMC Infect Dis. 2012;12:96. doi: 10.1186/1471-2334-12-96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Thakur H, Kaur N, Sareen D, Prabhakar N. Electrochemical determination of M. tuberculosis antigen based on Poly(3,4-ethylenedioxythiophene) and functionalized carbon nanotubes hybrid platform. Talanta. 2017;171:115–123. doi: 10.1016/j.talanta.2017.04.063. [DOI] [PubMed] [Google Scholar]
  • 13.Bai L, Chen Y, Bai Y, Chen Y, Zhou J, Huang A. Fullerene-doped polyaniline as new redox nanoprobe and catalyst in electrochemical aptasensor for ultrasensitive detection of Mycobacterium tuberculosis MPT64 antigen in human serum. Biomaterials. 2017;133:11–19. doi: 10.1016/j.biomaterials.2017.04.010. [DOI] [PubMed] [Google Scholar]
  • 14.Ngubane NA, Gresh L, Pym A, Rubin EJ, Khati M. Selection of RNA aptamers against the M. tuberculosis EsxG protein using surface plasmon resonance-based SELEX. Biochem Biophys Res Commun. 2014;449:114–119. doi: 10.1016/j.bbrc.2014.04.163. [DOI] [PubMed] [Google Scholar]
  • 15.Russell TM, Green LS, Rice T, Kruh-Garcia NA, Dobos K, De Groote MA. Potential of high-affinity, slow off-rate modified aptamer reagents for Mycobacterium tuberculosis proteins as tools for infection models and diagnostic applications. J Clin Microbiol. 2017;55:3072–3088. doi: 10.1128/JCM.00469-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Aimaiti R, Qin L, Cao T, Yang H, Wang J, Lu J. Identification and application of ssDNA aptamers against H(3)(7)Rv in the detection of Mycobacterium tuberculosis. Appl Microbiol Biotechnol. 2015;99:9073–9083. doi: 10.1007/s00253-015-6815-7. [DOI] [PubMed] [Google Scholar]
  • 17.Zhang X, Feng Y, Yao Q, He F. Selection of a new Mycobacterium tuberculosis H37Rv aptamer and its application in the construction of a SWCNT/aptamer/Au-IDE MSPQC H37Rv sensor. Biosens Bioelectron. 2017;98:261–266. doi: 10.1016/j.bios.2017.05.043. [DOI] [PubMed] [Google Scholar]
  • 18.Chen F, Zhou J, Luo F, Mohammed AB, Zhang XL. Aptamer from whole-bacterium SELEX as new therapeutic reagent against virulent Mycobacterium tuberculosis. Biochem Biophys Res Commun. 2007;357:743–748. doi: 10.1016/j.bbrc.2007.04.007. [DOI] [PubMed] [Google Scholar]
  • 19.Pan Q, Wang Q, Sun X, Xia X, Wu S, Luo F. Aptamer against mannose-capped lipoarabinomannan inhibits virulent Mycobacterium tuberculosis infection in mice and rhesus monkeys. Mol Ther. 2014;22:940–951. doi: 10.1038/mt.2014.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sun X, Pan Q, Yuan C, Wang Q, Tang XL, Ding K. A single ssDNA aptamer binding to ManLAM of BCG enhances immunoprotective effects against tuberculosis. J Am Chem Soc. 2016;138:11680–11689. doi: 10.1021/jacs.6b05357. [DOI] [PubMed] [Google Scholar]
  • 21.Pan Q, Yan J, Liu Q, Yuan C, Zhang XL. A single-stranded DNA aptamer against mannose-capped lipoarabinomannan enhances anti-tuberculosis activity of macrophages through downregulation of lipid-sensing nuclear receptor peroxisome proliferator-activated receptor gamma expression. Microbiol Immunol. 2017;61:92–102. doi: 10.1111/1348-0421.12470. [DOI] [PubMed] [Google Scholar]
  • 22.Baig IA, Moon JY, Lee SC, Ryoo SW, Yoon MY. Development of ssDNA aptamers as potent inhibitors of Mycobacterium tuberculosis acetohydroxyacid synthase. Biochim Biophys Acta. 2015;1854:1338–1350. doi: 10.1016/j.bbapap.2015.05.003. [DOI] [PubMed] [Google Scholar]
  • 23.Ranjbar R, Hafezi-Moghadam MS. Design and construction of a DNA origami drug delivery system based on MPT64 antibody aptamer for tuberculosis treatment. Electron Phys. 2016;8:1857–1864. doi: 10.19082/1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhang H, Ma X, Liu Y, Duan N, Wu S, Wang Z. Gold nanoparticles enhanced SERS aptasensor for the simultaneous detection of Salmonella typhimurium and Staphylococcus aureus. Biosens Bioelectron. 2015;74:872–877. doi: 10.1016/j.bios.2015.07.033. [DOI] [PubMed] [Google Scholar]
  • 25.Joshi R, Janagama H, Dwivedi HP, Senthil Kumar TM, Jaykus LA, Schefers J. Selection, characterization, and application of DNA aptamers for the capture and detection of Salmonella enterica serovars. Mol Cell Probes. 2009;23:20–28. doi: 10.1016/j.mcp.2008.10.006. [DOI] [PubMed] [Google Scholar]
  • 26.Ma X, Jiang Y, Jia F, Yu Y, Chen J, Wang Z. An aptamer-based electrochemical biosensor for the detection of Salmonella. J Microbiol Methods. 2014;98:94–98. doi: 10.1016/j.mimet.2014.01.003. [DOI] [PubMed] [Google Scholar]
  • 27.Bagheryan Z, Raoof JB, Golabi M, Turner AP, Beni V. Diazonium-based impedimetric aptasensor for the rapid label-free detection of Salmonella typhimurium in food sample. Biosens Bioelectron. 2016;80:566–573. doi: 10.1016/j.bios.2016.02.024. [DOI] [PubMed] [Google Scholar]
  • 28.Han SR, Lee SW. In vitro selection of RNA aptamer specific to Salmonella typhimurium. J Microbiol Biotechnol. 2013;23:878–884. doi: 10.4014/jmb.1212.12033. [DOI] [PubMed] [Google Scholar]
  • 29.Wu S, Duan N, Qiu Y, Li J, Wang Z. Colorimetric aptasensor for the detection of Salmonella enterica serovar typhimurium using ZnFe2O4-reduced graphene oxide nanostructures as an effective peroxidase mimetics. Int J Food Microbiol. 2017;261:42–48. doi: 10.1016/j.ijfoodmicro.2017.09.002. [DOI] [PubMed] [Google Scholar]
  • 30.Oh SY, Heo NS, Shukla S, Cho HJ, Vilian ATE, Kim J. Development of gold nanoparticle-aptamer-based LSPR sensing chips for the rapid detection of Salmonella typhimurium in pork meat. Sci Rep. 2017;7:10130. doi: 10.1038/s41598-017-10188-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Moon J, Kim G, Lee S, Park S. Identification of Salmonella Typhimurium-specific DNA aptamers developed using whole-cell SELEX and FACS analysis. J Microbiol Methods. 2013;95:162–166. doi: 10.1016/j.mimet.2013.08.005. [DOI] [PubMed] [Google Scholar]
  • 32.Duan N, Wu S, Chen X, Huang Y, Xia Y, Ma X. Selection and characterization of aptamers against Salmonella typhimurium using whole-bacterium Systemic Evolution of Ligands by Exponential Enrichment (SELEX) J Agric Food Chem. 2013;61:3229–3234. doi: 10.1021/jf400767d. [DOI] [PubMed] [Google Scholar]
  • 33.Lavu PS, Mondal B, Ramlal S, Murali HS, Batra HV. Selection and characterization of aptamers using a modified whole cell bacterium SELEX for the detection of Salmonella enterica serovar Typhimurium. ACS Comb Sci. 2016;18:292–301. doi: 10.1021/acscombsci.5b00123. [DOI] [PubMed] [Google Scholar]
  • 34.Yang M, Peng Z, Ning Y, Chen Y, Zhou Q, Deng L. Highly specific and cost-efficient detection of Salmonella Paratyphi A combining aptamers with single-walled carbon nanotubes. Sensors. 2013;13:6865–6881. doi: 10.3390/s130506865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bayrac C, Eyidogan F, Avni Oktem H. DNA aptamer-based colorimetric detection platform for Salmonella Enteritidis. Biosens Bioelectron. 2017;98:22–28. doi: 10.1016/j.bios.2017.06.029. [DOI] [PubMed] [Google Scholar]
  • 36.Duan N, Xu B, Wu S, Wang Z. Magnetic nanoparticles-based aptasensor using gold nanoparticles as colorimetric probes for the detection of Salmonella Typhimurium. Anal Sci. 2016;32:431–436. doi: 10.2116/analsci.32.431. [DOI] [PubMed] [Google Scholar]
  • 37.Wang QY, Kang YJ. Bioprobes based on aptamer and silica fluorescent nanoparticles for bacteria Salmonella Typhimurium detection. Nanoscale Res Lett. 2016;11:150. doi: 10.1186/s11671-016-1359-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hyeon JY, Chon JW, Choi IS, Park C, Kim DE, Seo KH. Development of RNA aptamers for detection of Salmonella Enteritidis. J Microbiol Methods. 2012;89:79–82. doi: 10.1016/j.mimet.2012.01.014. [DOI] [PubMed] [Google Scholar]
  • 39.Pan Q, Zhang XL, Wu HY, He PW, Wang F, Zhang MS. Aptamers that preferentially bind type IVB pili and inhibit human monocytic-cell invasion by Salmonella enterica serovar typhi. Antimicrob Agents Chemother. 2005;49:4052–4060. doi: 10.1128/AAC.49.10.4052-4060.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zelada-Guillen GA, Riu J, Duzgun A, Rius FX. Immediate detection of living bacteria at ultralow concentrations using a carbon nanotube based potentiometric aptasensor. Angew Chem Int Ed Engl. 2009;48:7334–7337. doi: 10.1002/anie.200902090. [DOI] [PubMed] [Google Scholar]
  • 41.Yeom JH, Lee B, Kim D, Lee JK, Kim S, Bae J. Gold nanoparticle-DNA aptamer conjugate-assisted delivery of antimicrobial peptide effectively eliminates intracellular Salmonella enterica serovar Typhimurium. Biomaterials. 2016;104:43–51. doi: 10.1016/j.biomaterials.2016.07.009. [DOI] [PubMed] [Google Scholar]
  • 42.DeGrasse JA. A single-stranded DNA aptamer that selectively binds to Staphylococcus aureus enterotoxin B. PLoS One. 2012;7:e33410. doi: 10.1371/journal.pone.0033410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Huang Y, Chen X, Duan N, Wu S, Wang Z, Wei X. Selection and characterization of DNA aptamers against Staphylococcus aureus enterotoxin C1. Food Chem. 2015;166:623–629. doi: 10.1016/j.foodchem.2014.06.039. [DOI] [PubMed] [Google Scholar]
  • 44.Hong KL, Battistella L, Salva AD, Williams RM, Sooter LJ. In vitro selection of single-stranded DNA molecular recognition elements against S. aureus alpha toxin and sensitive detection in human serum. Int J Mol Sci. 2015;16:2794–2809. doi: 10.3390/ijms16022794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ferreira IM, de Souza Lacerda CM, de Faria LS, Correa CR, de Andrade AS. Selection of peptidoglycan-specific aptamers for bacterial cells identification. Appl Biochem Biotechnol. 2014;174:2548–2556. doi: 10.1007/s12010-014-1206-6. [DOI] [PubMed] [Google Scholar]
  • 46.Stoltenburg R, Schubert T, Strehlitz B. In vitro selection and interaction studies of a DNA aptamer targeting protein A. PLoS One. 2015;10 doi: 10.1371/journal.pone.0134403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cao X, Li S, Chen L, Ding H, Xu H, Huang Y. Combining use of a panel of ssDNA aptamers in the detection of Staphylococcus aureus. Nucleic Acids Res. 2009;37:4621–4628. doi: 10.1093/nar/gkp489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Borsa BA, Tuna BG, Hernandez FJ, Hernandez LI, Bayramoglu G, Arica MY. Staphylococcus aureus detection in blood samples by silica nanoparticle-oligonucleotides conjugates. Biosens Bioelectron. 2016;86:27–32. doi: 10.1016/j.bios.2016.06.023. [DOI] [PubMed] [Google Scholar]
  • 49.Chang YC, Yang CY, Sun RL, Cheng YF, Kao WC, Yang PC. Rapid single cell detection of Staphylococcus aureus by aptamer-conjugated gold nanoparticles. Sci Rep. 2013;3:1863. doi: 10.1038/srep01863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang J, Wu H, Yang Y, Yan R, Zhao Y, Wang Y. Bacterial species-identifiable magnetic nanosystems for early sepsis diagnosis and extracorporeal photodynamic blood disinfection. Nanoscale. 2017;10:132–141. doi: 10.1039/c7nr06373c. [DOI] [PubMed] [Google Scholar]
  • 51.Wang K, Wu D, Chen Z, Zhang X, Yang X, Yang CJ. Inhibition of the superantigenic activities of Staphylococcal enterotoxin A by an aptamer antagonist. Toxicon. 2016;119:21–27. doi: 10.1016/j.toxicon.2016.05.006. [DOI] [PubMed] [Google Scholar]
  • 52.Vivekananda J, Salgado C, Millenbaugh NJ. DNA aptamers as a novel approach to neutralize Staphylococcus aureus alpha-toxin. Biochem Biophys Res Commun. 2014;444:433–438. doi: 10.1016/j.bbrc.2014.01.076. [DOI] [PubMed] [Google Scholar]
  • 53.Kavruk M, Celikbicak O, Ozalp VC, Borsa BA, Hernandez FJ, Bayramoglu G. Antibiotic loaded nanocapsules functionalized with aptamer gates for targeted destruction of pathogens. Chem Commun. 2015;51:8492–8495. doi: 10.1039/c5cc01869b. [DOI] [PubMed] [Google Scholar]
  • 54.Ocsoy I, Yusufbeyoglu S, Yilmaz V, McLamore ES, Ildiz N, Ulgen A. DNA aptamer functionalized gold nanostructures for molecular recognition and photothermal inactivation of methicillin-resistant Staphylococcus aureus. Colloids Surf B Biointerfaces. 2017;159:16–22. doi: 10.1016/j.colsurfb.2017.07.056. [DOI] [PubMed] [Google Scholar]
  • 55.Ohk SH, Koo OK, Sen T, Yamamoto CM, Bhunia AK. Antibody-aptamer functionalized fiber-optic biosensor for specific detection of Listeria monocytogenes from food. J Appl Microbiol. 2010;109:808–817. doi: 10.1111/j.1365-2672.2010.04709.x. [DOI] [PubMed] [Google Scholar]
  • 56.Bruno JG, Phillips T, Montez T, Garcia A, Sivils JC, Mayo MW. Development of a fluorescent enzyme-linked DNA aptamer-magnetic bead sandwich assay and portable fluorometer for sensitive and rapid listeria detection. J Fluoresc. 2015;25:173–183. doi: 10.1007/s10895-014-1495-8. [DOI] [PubMed] [Google Scholar]
  • 57.Suh SH, Jaykus LA. Nucleic acid aptamers for capture and detection of Listeria spp. J Biotechnol. 2013;167:454–461. doi: 10.1016/j.jbiotec.2013.07.027. [DOI] [PubMed] [Google Scholar]
  • 58.Suh SH, Dwivedi HP, Choi SJ, Jaykus LA. Selection and characterization of DNA aptamers specific for Listeria species. Anal Biochem. 2014;459:39–45. doi: 10.1016/j.ab.2014.05.006. [DOI] [PubMed] [Google Scholar]
  • 59.Lee SH, Ahn JY, Lee KA, Um HJ, Sekhon SS, Sun Park T. Analytical bioconjugates, aptamers, enable specific quantitative detection of Listeria monocytogenes. Biosens Bioelectron. 2015;68:272–280. doi: 10.1016/j.bios.2015.01.009. [DOI] [PubMed] [Google Scholar]
  • 60.Bruno JG, Chanpong J. Methods of producing competitive aptamer fret reagents and assays. 2009. http://www.freepatentsonline.com/y2009/0186342.html United States Patent Application 20090186342Available from URL:
  • 61.Khang J, Kim D, Chung KW, Lee JH. Chemiluminescent aptasensor capable of rapidly quantifying Escherichia coli O157:H7. Talanta. 2016;147:177–183. doi: 10.1016/j.talanta.2015.09.055. [DOI] [PubMed] [Google Scholar]
  • 62.Wu WH, Li M, Wang Y, Ouyang HX, Wang L, Li CX. Aptasensors for rapid detection of Escherichia coli O157:H7 and Salmonella typhimurium. Nanoscale Res Lett. 2012;7:658. doi: 10.1186/1556-276X-7-658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Amraee M, Oloomi M, Yavari A, Bouzari S. DNA aptamer identification and characterization for E. coli O157 detection using cell based SELEX method. Anal Biochem. 2017;536:36–44. doi: 10.1016/j.ab.2017.08.005. [DOI] [PubMed] [Google Scholar]
  • 64.Peng Z, Ling M, Ning Y, Deng L. Rapid fluorescent detection of Escherichia coli K88 based on DNA aptamer library as direct and specific reporter combined with immuno-magnetic separation. J Fluoresc. 2014;24:1159–1168. doi: 10.1007/s10895-014-1396-x. [DOI] [PubMed] [Google Scholar]
  • 65.Kim YS, Song MY, Jurng J, Kim BC. Isolation and characterization of DNA aptamers against Escherichia coli using a bacterial cell-systematic evolution of ligands by exponential enrichment approach. Anal Biochem. 2013;436:22–28. doi: 10.1016/j.ab.2013.01.014. [DOI] [PubMed] [Google Scholar]
  • 66.Chung J, Kang JS, Jurng JS, Jung JH, Kim BC. Fast and continuous microorganism detection using aptamer-conjugated fluorescent nanoparticles on an optofluidic platform. Biosens Bioelectron. 2015;67:303–308. doi: 10.1016/j.bios.2014.08.039. [DOI] [PubMed] [Google Scholar]
  • 67.Jin B, Wang S, Lin M, Jin Y, Zhang S, Cui X. Upconversion nanoparticles based FRET aptasensor for rapid and ultrasenstive bacteria detection. Biosens Bioelectron. 2017;90:525–533. doi: 10.1016/j.bios.2016.10.029. [DOI] [PubMed] [Google Scholar]
  • 68.Guo Y, Wang Y, Liu S, Yu J, Wang H, Wang Y. Label-free and highly sensitive electrochemical detection of E. coli based on rolling circle amplifications coupled peroxidase-mimicking DNAzyme amplification. Biosens Bioelectron. 2016;75:315–319. doi: 10.1016/j.bios.2015.08.031. [DOI] [PubMed] [Google Scholar]
  • 69.Dua P, Ren S, Lee SW, Kim JK, Shin HS, Jeong OC. Cell-SELEX based identification of an RNA aptamer for Escherichia coli and its use in various detection formats. Mol Cells. 2016;39:807–813. doi: 10.14348/molcells.2016.0167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Renders M, Miller E, Lam CH, Perrin DM. Whole cell-SELEX of aptamers with a tyrosine-like side chain against live bacteria. Org Biomol Chem. 2017;15:1980–1989. doi: 10.1039/c6ob02451c. [DOI] [PubMed] [Google Scholar]
  • 71.Bruno JG, Carrillo MP, Phillips T. In vitro antibacterial effects of antilipopolysaccharide DNA aptamer-C1qrs complexes. Folia Microbiol. 2008;53:295–302. doi: 10.1007/s12223-008-0046-6. [DOI] [PubMed] [Google Scholar]
  • 72.Fatin MF, Ruslinda AR, Arshad MK, Tee KK, Ayub RM, Hashim U. HIV-1 Tat biosensor: current development and trends for early detection strategies. Biosens Bioelectron. 2016;78:358–366. doi: 10.1016/j.bios.2015.11.067. [DOI] [PubMed] [Google Scholar]
  • 73.Yamamoto R, Katahira M, Nishikawa S, Baba T, Taira K, Kumar PK. A novel RNA motif that binds efficiently and specifically to the Tat protein of HIV and inhibits the trans-activation by Tat of transcription in vitro and in vivo. Genes Cells. 2000;5:371–388. doi: 10.1046/j.1365-2443.2000.00330.x. [DOI] [PubMed] [Google Scholar]
  • 74.Minunni M, Tombelli S, Gullotto A, Luzi E, Mascini M. Development of biosensors with aptamers as bio-recognition element: the case of HIV-1 Tat protein. Biosens Bioelectron. 2004;20:1149–1156. doi: 10.1016/j.bios.2004.03.037. [DOI] [PubMed] [Google Scholar]
  • 75.Tombelli S, Minunni M, Luzi E, Mascini M. Aptamer-based biosensors for the detection of HIV-1 Tat protein. Bioelectrochemistry. 2005;67:135–141. doi: 10.1016/j.bioelechem.2004.04.011. [DOI] [PubMed] [Google Scholar]
  • 76.Rahim Ruslinda A, Tanabe K, Ibori S, Wang X, Kawarada H. Effects of diamond-FET-based RNA aptamer sensing for detection of real sample of HIV-1 Tat protein. Biosens Bioelectron. 2013;40:277–282. doi: 10.1016/j.bios.2012.07.048. [DOI] [PubMed] [Google Scholar]
  • 77.Sayer N, Ibrahim J, Turner K, Tahiri-Alaoui A, James W. Structural characterization of a 2′F-RNA aptamer that binds a HIV-1 SU glycoprotein, gp120. Biochem Biophys Res Commun. 2002;293:924–931. doi: 10.1016/S0006-291X(02)00308-X. [DOI] [PubMed] [Google Scholar]
  • 78.Dey AK, Griffiths C, Lea SM, James W. Structural characterization of an anti-gp120 RNA aptamer that neutralizes R5 strains of HIV-1. RNA. 2005;11:873–884. doi: 10.1261/rna.7205405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cohen C, Forzan M, Sproat B, Pantophlet R, McGowan I, Burton D. An aptamer that neutralizes R5 strains of HIV-1 binds to core residues of gp120 in the CCR5 binding site. Virology. 2008;381:46–54. doi: 10.1016/j.virol.2008.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhou J, Swiderski P, Li H, Zhang J, Neff CP, Akkina R, Rossi JJ. Selection, characterization and application of new RNA HIV gp 120 aptamers for facile delivery of Dicer substrate siRNAs into HIV infected cells. Nucleic Acids Res. 2009;37:3094–3109. doi: 10.1093/nar/gkp185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Dey AK, Khati M, Tang M, Wyatt R, Lea SM, James W. An aptamer that neutralizes R5 strains of human immunodeficiency virus type 1 blocks gp120-CCR5 interaction. J Virol. 2005;79:13806–13810. doi: 10.1128/JVI.79.21.13806-13810.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lopes de Campos WR, Chirwa N, London G, Rotherham LS, Morris L, Mayosi BM. HIV-1 subtype C unproductively infects human cardiomyocytes in vitro and induces apoptosis mitigated by an anti-Gp120 aptamer. PLoS One. 2014;9 doi: 10.1371/journal.pone.0110930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Khati M, Schuman M, Ibrahim J, Sattentau Q, Gordon S, James W. Neutralization of infectivity of diverse R5 clinical isolates of human immunodeficiency virus type 1 by gp120-binding 2′F-RNA aptamers. J Virol. 2003;77:12692–12698. doi: 10.1128/JVI.77.23.12692-12698.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Neff CP, Zhou J, Remling L, Kuruvilla J, Zhang J, Li H. An aptamer-siRNA chimera suppresses HIV-1 viral loads and protects from helper CD4(+) T cell decline in humanized mice. Sci Transl Med. 2011;3:66ra6. doi: 10.1126/scitranslmed.3001581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zhou J, Neff CP, Swiderski P, Li H, Smith DD, Aboellail T. Functional in vivo delivery of multiplexed anti-HIV-1 siRNAs via a chemically synthesized aptamer with a sticky bridge. Mol Ther. 2013;21:192–200. doi: 10.1038/mt.2012.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Prokofjeva M, Tsvetkov V, Basmanov D, Varizhuk A, Lagarkova M, Smirnov I. Anti-HIV activities of intramolecular G4 and non-G4 oligonucleotides. Nucleic Acid Ther. 2017;27:56–66. doi: 10.1089/nat.2016.0624. [DOI] [PubMed] [Google Scholar]
  • 87.Aeksiri N, Songtawee N, Gleeson MP, Hannongbua S, Choowongkomon K. Insight into HIV-1 reverse transcriptase-aptamer interaction from molecular dynamics simulations. J Mol Model. 2014;20:2380. doi: 10.1007/s00894-014-2380-8. [DOI] [PubMed] [Google Scholar]
  • 88.Tuerk C, MacDougal S, Gold L. RNA pseudoknots that inhibit human immunodeficiency virus type 1 reverse transcriptase. Proc Natl Acad Sci USA. 1992;89:6988–6992. doi: 10.1073/pnas.89.15.6988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Schneider DJ, Feigon J, Hostomsky Z, Gold L. High-affinity ssDNA inhibitors of the reverse transcriptase of type 1 human immunodeficiency virus. Biochemistry. 1995;34:9599–9610. doi: 10.1021/bi00029a037. [DOI] [PubMed] [Google Scholar]
  • 90.Mosing RK, Mendonsa SD, Bowser MT. Capillary electrophoresis-SELEX selection of aptamers with affinity for HIV-1 reverse transcriptase. Anal Chem. 2005;77:6107–6112. doi: 10.1021/ac050836q. [DOI] [PubMed] [Google Scholar]
  • 91.Somasunderam A, Ferguson MR, Rojo DR, Thiviyanathan V, Li X, O'Brien WA. Combinatorial selection, inhibition, and antiviral activity of DNA thioaptamers targeting the RNase H domain of HIV-1 reverse transcriptase. Biochemistry. 2005;44:10388–10395. doi: 10.1021/bi0507074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kissel JD, Held DM, Hardy RW, Burke DH. Single-stranded DNA aptamer RT1t49 inhibits RT polymerase and RNase H functions of HIV type 1, HIV type 2, and SIVCPZ RTs. AIDS Res Hum Retrovir. 2007;23:699–708. doi: 10.1089/aid.2006.0262. [DOI] [PubMed] [Google Scholar]
  • 93.DeStefano JJ, Nair GR. Novel aptamer inhibitors of human immunodeficiency virus reverse transcriptase. Oligonucleotides. 2008;18:133–144. doi: 10.1089/oli.2008.0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lai YT, DeStefano JJ. DNA aptamers to human immunodeficiency virus reverse transcriptase selected by a primer-free SELEX method: characterization and comparison with other aptamers. Nucleic Acid Ther. 2012;22:162–176. doi: 10.1089/nat.2011.0327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Alves Ferreira-Bravo I, Cozens C, Holliger P, DeStefano JJ. Selection of 2′-deoxy-2′-fluoroarabinonucleotide (FANA) aptamers that bind HIV-1 reverse transcriptase with picomolar affinity. Nucleic Acids Res. 2015;43:9587–9599. doi: 10.1093/nar/gkv1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sanchez-Luque FJ, Stich M, Manrubia S, Briones C, Berzal-Herranz A. Efficient HIV-1 inhibition by a 16 nt-long RNA aptamer designed by combining in vitro selection and in silico optimisation strategies. Sci Rep. 2014;4:6242. doi: 10.1038/srep06242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Boiziau C, Dausse E, Yurchenko L, Toulme JJ. DNA aptamers selected against the HIV-1 trans-activation-responsive RNA element form RNA-DNA kissing complexes. J Biol Chem. 1999;274:12730–12737. doi: 10.1074/jbc.274.18.12730. [DOI] [PubMed] [Google Scholar]
  • 98.Jing N, Hogan ME. Structure-activity of tetrad-forming oligonucleotides as a potent anti-HIV therapeutic drug. J Biol Chem. 1998;273:34992–34999. doi: 10.1074/jbc.273.52.34992. [DOI] [PubMed] [Google Scholar]
  • 99.De Soultrait VR, Lozach PY, Altmeyer R, Tarrago-Litvak L, Litvak S, Andreola ML. DNA aptamers derived from HIV-1 RNase H inhibitors are strong anti-integrase agents. J Mol Biol. 2002;324:195–203. doi: 10.1016/s0022-2836(02)01064-1. [DOI] [PubMed] [Google Scholar]
  • 100.Phan AT, Kuryavyi V, Ma JB, Faure A, Andreola ML, Patel DJ. An interlocked dimeric parallel-stranded DNA quadruplex: a potent inhibitor of HIV-1 integrase. Proc Natl Acad Sci USA. 2005;102:634–639. doi: 10.1073/pnas.0406278102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Faure-Perraud A, Metifiot M, Reigadas S, Recordon-Pinson P, Parissi V, Ventura M. The guanine-quadruplex aptamer 93del inhibits HIV-1 replication ex vivo by interfering with viral entry, reverse transcription and integration. Antivir Ther. 2011;16:383–394. doi: 10.3851/IMP1756. [DOI] [PubMed] [Google Scholar]
  • 102.Esposito V, Pirone L, Mayol L, Pedone E, Virgilio A, Galeone A. Exploring the binding of d(GGGT)4 to the HIV-1 integrase: an approach to investigate G-quadruplex aptamer/target protein interactions. Biochimie. 2016;127:19–22. doi: 10.1016/j.biochi.2016.04.013. [DOI] [PubMed] [Google Scholar]
  • 103.Kim SJ, Kim MY, Lee JH, You JC, Jeong S. Selection and stabilization of the RNA aptamers against the human immunodeficiency virus type-1 nucleocapsid protein. Biochem Biophys Res Commun. 2002;291:925–931. doi: 10.1006/bbrc.2002.6521. [DOI] [PubMed] [Google Scholar]
  • 104.Ramalingam D, Duclair S, Datta SA, Ellington A, Rein A, Prasad VR. RNA aptamers directed to human immunodeficiency virus type 1 Gag polyprotein bind to the matrix and nucleocapsid domains and inhibit virus production. J Virol. 2011;85:305–314. doi: 10.1128/JVI.02626-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Jensen KB, Green L, MacDougal-Waugh S, Tuerk C. Characterization of an in vitro-selected RNA ligand to the HIV-1 Rev protein. J Mol Biol. 1994;235:237–247. doi: 10.1016/s0022-2836(05)80030-0. [DOI] [PubMed] [Google Scholar]
  • 106.Pang KM, Castanotto D, Li H, Scherer L, Rossi JJ. Incorporation of aptamers in the terminal loop of shRNAs yields an effective and novel combinatorial targeting strategy. Nucleic Acids Res. 2017 doi: 10.1093/nar/gkx980. doi: 10.1093/nar/gkx980. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Alavian SM, Haghbin H. Relative importance of hepatitis B and C viruses in hepatocellular carcinoma in EMRO countries and the Middle East: a systematic review. Hepat Mon. 2016;16:e35106. doi: 10.5812/hepatmon.35106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Chen F, Hu Y, Li D, Chen H, Zhang XL. CS-SELEX generates high-affinity ssDNA aptamers as molecular probes for hepatitis C virus envelope glycoprotein E2. PLoS One. 2009;4:e8142. doi: 10.1371/journal.pone.0008142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lee S, Kim YS, Jo M, Jin M, Lee DK, Kim S. Chip-based detection of hepatitis C virus using RNA aptamers that specifically bind to HCV core antigen. Biochem Biophys Res Commun. 2007;358:47–52. doi: 10.1016/j.bbrc.2007.04.057. [DOI] [PubMed] [Google Scholar]
  • 110.Shi S, Yu X, Gao Y, Xue B, Wu X, Wang X. Inhibition of hepatitis C virus production by aptamers against the core protein. J Virol. 2014;88:1990–1999. doi: 10.1128/JVI.03312-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ghanbari K, Roushani M, Azadbakht A. Ultra-sensitive aptasensor based on a GQD nanocomposite for detection of hepatitis C virus core antigen. Anal Biochem. 2017;534:64–69. doi: 10.1016/j.ab.2017.07.016. [DOI] [PubMed] [Google Scholar]
  • 112.Kumar PK, Machida K, Urvil PT, Kakiuchi N, Vishnuvardhan D, Shimotohno K. Isolation of RNA aptamers specific to the NS3 protein of hepatitis C virus from a pool of completely random RNA. Virology. 1997;237:270–282. doi: 10.1006/viro.1997.8773. [DOI] [PubMed] [Google Scholar]
  • 113.Fukuda K, Vishnuvardhan D, Sekiya S, Hwang J, Kakiuchi N, Taira K. Isolation and characterization of RNA aptamers specific for the hepatitis C virus nonstructural protein 3 protease. Eur J Biochem. 2000;267:3685–3694. doi: 10.1046/j.1432-1327.2000.01400.x. [DOI] [PubMed] [Google Scholar]
  • 114.Fukuda K, Sekiya S, Kikuchi K, Funaji K, Kuno A, Hasegawa T. Construction of the dual-functional RNA ligand against HCV NS3 protease and helicase. Nucleic Acids Res Suppl. 2001;1:147–148. doi: 10.1093/nass/1.1.147. [DOI] [PubMed] [Google Scholar]
  • 115.Fukuda K, Umehara T, Sekiya S, Kunio K, Hasegawa T, Nishikawa S. An RNA ligand inhibits hepatitis C virus NS3 protease and helicase activities. Biochem Biophys Res Commun. 2004;325:670–675. doi: 10.1016/j.bbrc.2004.10.089. [DOI] [PubMed] [Google Scholar]
  • 116.Nishikawa F, Funaji K, Fukuda K, Nishikawa S. In vitro selection of RNA aptamers against the HCV NS3 helicase domain. Oligonucleotides. 2004;14:114–129. doi: 10.1089/1545457041526335. [DOI] [PubMed] [Google Scholar]
  • 117.Kikuchi K, Umehara T, Fukuda K, Kuno A, Hasegawa T, Nishikawa S. A hepatitis C virus (HCV) internal ribosome entry site (IRES) domain III-IV-targeted aptamer inhibits translation by binding to an apical loop of domain IIId. Nucleic Acids Res. 2005;33:683–692. doi: 10.1093/nar/gki215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Romero-Lopez C, Barroso-delJesus A, Puerta-Fernandez E, Berzal-Herranz A. Interfering with hepatitis C virus IRES activity using RNA molecules identified by a novel in vitro selection method. Biol Chem. 2005;386:183–190. doi: 10.1515/BC.2005.023. [DOI] [PubMed] [Google Scholar]
  • 119.Romero-Lopez C, Diaz-Gonzalez R, Berzal-Herranz A. Inhibition of hepatitis C virus internal ribosome entry site-mediated translation by an RNA targeting the conserved IIIf domain. Cell Mol Life Sci. 2007;64:2994–3006. doi: 10.1007/s00018-007-7345-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Romero-Lopez C, Lahlali T, Berzal-Herranz B, Berzal-Herranz A. Development of optimized inhibitor RNAs allowing multisite-targeting of the HCV genome. Molecules. 2017;22::E861. doi: 10.3390/molecules22050861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bellecave P, Andreola ML, Ventura M, Tarrago-Litvak L, Litvak S, Astier-Gin T. Selection of DNA aptamers that bind the RNA-dependent RNA polymerase of hepatitis C virus and inhibit viral RNA synthesis in vitro. Oligonucleotides. 2003;13:455–463. doi: 10.1089/154545703322860771. [DOI] [PubMed] [Google Scholar]
  • 122.Bellecave P, Cazenave C, Rumi J, Staedel C, Cosnefroy O, Andreola ML. Inhibition of hepatitis C virus (HCV) RNA polymerase by DNA aptamers: mechanism of inhibition of in vitro RNA synthesis and effect on HCV-infected cells. Antimicrob Agents Chemother. 2008;52:2097–2110. doi: 10.1128/AAC.01227-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Jones LA, Clancy LE, Rawlinson WD, White PA. High-affinity aptamers to subtype 3a hepatitis C virus polymerase display genotypic specificity. Antimicrob Agents Chemother. 2006;50:3019–3027. doi: 10.1128/AAC.01603-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Lee CH, Lee YJ, Kim JH, Lim JH, Kim JH, Han W. Inhibition of hepatitis C virus (HCV) replication by specific RNA aptamers against HCV NS5B RNA replicase. J Virol. 2013;87:7064–7074. doi: 10.1128/JVI.00405-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lee CH, Lee SH, Kim JH, Noh YH, Noh GJ, Lee SW. Pharmacokinetics of a cholesterol-conjugated aptamer against the hepatitis C virus (HCV) NS5B protein. Mol Ther Nucleic Acids. 2015;4:e254. doi: 10.1038/mtna.2015.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hung LY, Huang TB, Tsai YC, Yeh CS, Lei HY, Lee GB. A microfluidic immunomagnetic bead-based system for the rapid detection of influenza infections: from purified virus particles to clinical specimens. Biomed Microdevices. 2013;15:539–551. doi: 10.1007/s10544-013-9753-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Gopinath SC, Tang TH, Chen Y, Citartan M, Tominaga J, Lakshmipriya T. Sensing strategies for influenza surveillance. Biosens Bioelectron. 2014;61:357–369. doi: 10.1016/j.bios.2014.05.024. [DOI] [PubMed] [Google Scholar]
  • 128.Jeon SH, Kayhan B, Ben-Yedidia T, Arnon R. A DNA aptamer prevents influenza infection by blocking the receptor binding region of the viral hemagglutinin. J Biol Chem. 2004;279:48410–48419. doi: 10.1074/jbc.M409059200. [DOI] [PubMed] [Google Scholar]
  • 129.Misono TS, Kumar PK. Selection of RNA aptamers against human influenza virus hemagglutinin using surface plasmon resonance. Anal Biochem. 2005;342:312–317. doi: 10.1016/j.ab.2005.04.013. [DOI] [PubMed] [Google Scholar]
  • 130.Gopinath SC, Misono TS, Kawasaki K, Mizuno T, Imai M, Odagiri T. An RNA aptamer that distinguishes between closely related human influenza viruses and inhibits haemagglutinin-mediated membrane fusion. J Gen Virol. 2006;87:479–487. doi: 10.1099/vir.0.81508-0. [DOI] [PubMed] [Google Scholar]
  • 131.Cheng C, Dong J, Yao L, Chen A, Jia R, Huan L. Potent inhibition of human influenza H5N1 virus by oligonucleotides derived by SELEX. Biochem Biophys Res Commun. 2008;366:670–674. doi: 10.1016/j.bbrc.2007.11.183. [DOI] [PubMed] [Google Scholar]
  • 132.Choi SK, Lee C, Lee KS, Choe SY, Mo IP, Seong RH. DNA aptamers against the receptor binding region of hemagglutinin prevent avian influenza viral infection. Mol Cells. 2011;32:527–533. doi: 10.1007/s10059-011-0156-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Park SY, Kim S, Yoon H, Kim KB, Kalme SS, Oh S. Selection of an antiviral RNA aptamer against hemagglutinin of the subtype H5 avian influenza virus. Nucleic Acid Ther. 2011;21:395–402. doi: 10.1089/nat.2011.0321. [DOI] [PubMed] [Google Scholar]
  • 134.Gopinath SC, Kumar PK. Aptamers that bind to the hemagglutinin of the recent pandemic influenza virus H1N1 and efficiently inhibit agglutination. Acta Biomater. 2013;9:8932–8941. doi: 10.1016/j.actbio.2013.06.016. [DOI] [PubMed] [Google Scholar]
  • 135.Wongphatcharachai M, Wang P, Enomoto S, Webby RJ, Gramer MR, Amonsin A. Neutralizing DNA aptamers against swine influenza H3N2 viruses. J Clin Microbiol. 2013;51:46–54. doi: 10.1128/JCM.02118-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wang R, Zhao J, Jiang T, Kwon YM, Lu H, Jiao P. Selection and characterization of DNA aptamers for use in detection of avian influenza virus H5N1. J Virol Methods. 2013;189:362–369. doi: 10.1016/j.jviromet.2013.03.006. [DOI] [PubMed] [Google Scholar]
  • 137.Suenaga E, Kumar PK. An aptamer that binds efficiently to the hemagglutinins of highly pathogenic avian influenza viruses (H5N1 and H7N7) and inhibits hemagglutinin-glycan interactions. Acta Biomater. 2014;10:1314–1323. doi: 10.1016/j.actbio.2013.12.034. [DOI] [PubMed] [Google Scholar]
  • 138.Woo HM, Lee JM, Yim S, Jeong YJ. Isolation of single-stranded DNA aptamers that distinguish influenza virus hemagglutinin subtype H1 from H5. PLoS One. 2015;10 doi: 10.1371/journal.pone.0125060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Zhang Y, Yu Z, Jiang F, Fu P, Shen J, Wu W. Two DNA aptamers against avian influenza H9N2 virus prevent viral infection in cells. PLoS One. 2015;10 doi: 10.1371/journal.pone.0123060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bai H, Wang R, Hargis B, Lu H, Li Y. A SPR aptasensor for detection of avian influenza virus H5N1. Sensors. 2012;12:12506–12518. doi: 10.3390/s120912506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Karash S, Wang R, Kelso L, Lu H, Huang TJ, Li Y. Rapid detection of avian influenza virus H5N1 in chicken tracheal samples using an impedance aptasensor with gold nanoparticles for signal amplification. J Virol Methods. 2016;236:147–156. doi: 10.1016/j.jviromet.2016.07.018. [DOI] [PubMed] [Google Scholar]
  • 142.Lai HC, Wang CH, Liou TM, Lee GB. Influenza A virus-specific aptamers screened by using an integrated microfluidic system. Lab Chip. 2014;14:2002–2013. doi: 10.1039/c4lc00187g. [DOI] [PubMed] [Google Scholar]
  • 143.Tseng YT, Wang CH, Chang CP, Lee GB. Integrated microfluidic system for rapid detection of influenza H1N1 virus using a sandwich-based aptamer assay. Biosens Bioelectron. 2016;82:105–111. doi: 10.1016/j.bios.2016.03.073. [DOI] [PubMed] [Google Scholar]
  • 144.Nguyen VT, Seo HB, Kim BC, Kim SK, Song CS, Gu MB. Highly sensitive sandwich-type SPR based detection of whole H5Nx viruses using a pair of aptamers. Biosens Bioelectron. 2016;86:293–300. doi: 10.1016/j.bios.2016.06.064. [DOI] [PubMed] [Google Scholar]
  • 145.Kwon HM, Lee KH, Han BW, Han MR, Kim DH, Kim DE. An RNA aptamer that specifically binds to the glycosylated hemagglutinin of avian influenza virus and suppresses viral infection in cells. PLoS One. 2014;9:e97574. doi: 10.1371/journal.pone.0097574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yuan S, Zhang N, Singh K, Shuai H, Chu H, Zhou J. Cross-protection of influenza A virus infection by a DNA aptamer targeting the PA endonuclease domain. Antimicrob Agents Chemother. 2015;59:4082–4093. doi: 10.1128/AAC.00306-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Liu J, Yang Y, Hu B, Ma ZY, Huang HP, Yu Y. Development of HBsAg-binding aptamers that bind HepG2.2.15 cells via HBV surface antigen. Virol Sin. 2010;25:27–35. doi: 10.1007/s12250-010-3091-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Feng H, Beck J, Nassal M, Hu KH. A SELEX-screened aptamer of human hepatitis B virus RNA encapsidation signal suppresses viral replication. PLoS One. 2011;6:e27862. doi: 10.1371/journal.pone.0027862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Zhang Z, Zhang J, Pei X, Zhang Q, Lu B, Zhang X. An aptamer targets HBV core protein and suppresses HBV replication in HepG2.2.15 cells. Int J Mol Med. 2014;34:1423–1429. doi: 10.3892/ijmm.2014.1908. [DOI] [PubMed] [Google Scholar]
  • 150.Orabi A, Bieringer M, Geerlof A, Bruss V. An aptamer against the matrix binding domain on the hepatitis B virus capsid impairs virion formation. J Virol. 2015;89:9281–9287. doi: 10.1128/JVI.00466-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Nicol C, Bunka DH, Blair GE, Stonehouse NJ. Effects of single nucleotide changes on the binding and activity of RNA aptamers to human papillomavirus 16 E7 oncoprotein. Biochem Biophys Res Commun. 2011;405:417–421. doi: 10.1016/j.bbrc.2011.01.044. [DOI] [PubMed] [Google Scholar]
  • 152.Toscano-Garibay JD, Benitez-Hess ML, Alvarez-Salas LM. Isolation and characterization of an RNA aptamer for the HPV-16 E7 oncoprotein. Arch Med Res. 2011;42:88–96. doi: 10.1016/j.arcmed.2011.02.005. [DOI] [PubMed] [Google Scholar]
  • 153.Nicol C, Cesur O, Forrest S, Belyaeva TA, Bunka DH, Blair GE. An RNA aptamer provides a novel approach for the induction of apoptosis by targeting the HPV16 E7 oncoprotein. PLoS One. 2013;8:e64781. doi: 10.1371/journal.pone.0064781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Belyaeva TA, Nicol C, Cesur O, Trave G, Blair GE, Stonehouse NJ. An RNA aptamer targets the PDZ-binding motif of the HPV16 E6 oncoprotein. Cancers. 2014;6:1553–1569. doi: 10.3390/cancers6031553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Shum KT, Tanner JA. Differential inhibitory activities and stabilisation of DNA aptamers against the SARS coronavirus helicase. Chembiochem. 2008;9:3037–3045. doi: 10.1002/cbic.200800491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Chen HL, Hsiao WH, Lee HC, Wu SC, Cheng JW. Selection and characterization of DNA aptamers targeting all four serotypes of dengue viruses. PLoS One. 2015;10 doi: 10.1371/journal.pone.0131240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Liang HR, Hu GQ, Li L, Gao YW, Yang ST, Xia XZ. Aptamers targeting rabies virus-infected cells inhibit street rabies virus in vivo. Int Immunopharmacol. 2014;21:432–438. doi: 10.1016/j.intimp.2014.03.020. [DOI] [PubMed] [Google Scholar]
  • 158.Liang HR, Hu GQ, Xue XH, Li L, Zheng XX, Gao YW. Selection of an aptamer against rabies virus: a new class of molecules with antiviral activity. Virus Res. 2014;184:7–13. doi: 10.1016/j.virusres.2014.01.021. [DOI] [PubMed] [Google Scholar]
  • 159.Piro B, Shi S, Reisberg S, Noel V, Anquetin G. Comparison of electrochemical immunosensors and aptasensors for detection of small organic molecules in environment. Biosensors. 2016;6:E7. doi: 10.3390/bios6010007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Duan N, Wu S, Yu Y, Ma X, Xia Y, Chen X. A dual-color flow cytometry protocol for the simultaneous detection of Vibrio parahaemolyticus and Salmonella typhimurium using aptamer conjugated quantum dots as labels. Anal Chim Acta. 2013;804:151–158. doi: 10.1016/j.aca.2013.09.047. [DOI] [PubMed] [Google Scholar]
  • 161.Kurt H, Yuce M, Hussain B, Budak H. Dual-excitation upconverting nanoparticle and quantum dot aptasensor for multiplexed food pathogen detection. Biosens Bioelectron. 2016;81:280–286. doi: 10.1016/j.bios.2016.03.005. [DOI] [PubMed] [Google Scholar]
  • 162.Liu J, Cheng J, Zhang Y. Upconversion nanoparticle based LRET system for sensitive detection of MRSA DNA sequence. Biosens Bioelectron. 2013;43:252–256. doi: 10.1016/j.bios.2012.12.026. [DOI] [PubMed] [Google Scholar]
  • 163.Wang L, Liu X, Hu X, Song S, Fan C. Unmodified gold nanoparticles as a colorimetric probe for potassium DNA aptamers. Chem Commun. 2006;36:3780–3782. doi: 10.1039/b607448k. [DOI] [PubMed] [Google Scholar]
  • 164.Bruno JG. Application of DNA aptamers and quantum dots to lateral flow test strips for detection of foodborne pathogens with improved sensitivity versus colloidal gold. Pathogens. 2014;3:341–355. doi: 10.3390/pathogens3020341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Zelada-Guillen GA, Bhosale SV, Riu J, Rius FX. Real-time potentiometric detection of bacteria in complex samples. Anal Chem. 2010;82:9254–9260. doi: 10.1021/ac101739b. [DOI] [PubMed] [Google Scholar]
  • 166.Zelada-Guillen GA, Sebastian-Avila JL, Blondeau P, Riu J, Rius FX. Label-free detection of Staphylococcus aureus in skin using real-time potentiometric biosensors based on carbon nanotubes and aptamers. Biosens Bioelectron. 2012;31:226–232. doi: 10.1016/j.bios.2011.10.021. [DOI] [PubMed] [Google Scholar]
  • 167.Hernandez R, Valles C, Benito AM, Maser WK, Rius FX, Riu J. Graphene-based potentiometric biosensor for the immediate detection of living bacteria. Biosens Bioelectron. 2014;54:553–557. doi: 10.1016/j.bios.2013.11.053. [DOI] [PubMed] [Google Scholar]
  • 168.Gronewold TM, Baumgartner A, Hierer J, Sierra S, Blind M, Schafer F. Kinetic binding analysis of aptamers targeting HIV-1 proteins by a combination of a microbalance array and mass spectrometry (MAMS) J Proteome Res. 2009;8:3568–3577. doi: 10.1021/pr900265r. [DOI] [PubMed] [Google Scholar]
  • 169.Wang R, Li Y. Hydrogel based QCM aptasensor for detection of avian influenza virus. Biosens Bioelectron. 2013;42:148–155. doi: 10.1016/j.bios.2012.10.038. [DOI] [PubMed] [Google Scholar]
  • 170.Dougan H, Lyster DM, Vo CV, Stafford A, Weitz JI, Hobbs JB. Extending the lifetime of anticoagulant oligodeoxynucleotide aptamers in blood. Nucl Med Biol. 2000;27:289–297. doi: 10.1016/s0969-8051(99)00103-1. [DOI] [PubMed] [Google Scholar]
  • 171.Fine SL, Martin DF, Kirkpatrick P. Pegaptanib sodium. Nat Rev Drug Discov. 2005;4:187–188. doi: 10.1038/nrd1677. [DOI] [PubMed] [Google Scholar]
  • 172.Chan MY, Cohen MG, Dyke CK, Myles SK, Aberle LG, Lin M. Phase 1b randomized study of antidote-controlled modulation of factor IXa activity in patients with stable coronary artery disease. Circulation. 2008;117:2865–2874. doi: 10.1161/CIRCULATIONAHA.107.745687. [DOI] [PubMed] [Google Scholar]
  • 173.Diener JL, Daniel Lagasse HA, Duerschmied D, Merhi Y, Tanguay JF, Hutabarat R. Inhibition of von Willebrand factor-mediated platelet activation and thrombosis by the anti-von Willebrand factor A1-domain aptamer ARC1779. J Thromb Haemost. 2009;7:1155–1162. doi: 10.1111/j.1538-7836.2009.03459.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Gao S, Zheng X, Jiao B, Wang L. Post-SELEX optimization of aptamers. Anal Bioanal Chem. 2016;408:4567–4573. doi: 10.1007/s00216-016-9556-2. [DOI] [PubMed] [Google Scholar]
  • 175.Karlsen KK, Wengel J. Locked nucleic acid and aptamers. Nucleic Acid Ther. 2012;22:366–370. doi: 10.1089/nat.2012.0382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Shigdar S, Macdonald J, O'Connor M, Wang T, Xiang D, Al Shamaileh H. Aptamers as theranostic agents: modifications, serum stability and functionalisation. Sensors. 2013;13:13624–13637. doi: 10.3390/s131013624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Heo K, Min SW, Sung HJ, Kim HG, Kim HJ, Kim YH. An aptamer-antibody complex (oligobody) as a novel delivery platform for targeted cancer therapies. J Control Release. 2016;229:1–9. doi: 10.1016/j.jconrel.2016.03.006. [DOI] [PubMed] [Google Scholar]
  • 178.Ara MN, Matsuda T, Hyodo M, Sakurai Y, Hatakeyama H, Ohga N. An aptamer ligand based liposomal nanocarrier system that targets tumor endothelial cells. Biomaterials. 2014;35:7110–7120. doi: 10.1016/j.biomaterials.2014.04.087. [DOI] [PubMed] [Google Scholar]
  • 179.Liao J, Liu B, Liu J, Zhang J, Chen K, Liu H. Cell-specific aptamers and their conjugation with nanomaterials for targeted drug delivery. Expert Opin Drug Deliv. 2015;12:493–506. doi: 10.1517/17425247.2015.966681. [DOI] [PubMed] [Google Scholar]
  • 180.Swierczewska M, Lee KC, Lee S. What is the future of PEGylated therapies? Expert Opin Emerg Drugs. 2015;20:531–536. doi: 10.1517/14728214.2015.1113254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Nimjee SM, White RR, Becker RC, Sullenger BA. Aptamers as therapeutics. Annu Rev Pharmacol Toxicol. 2017;57:61–79. doi: 10.1146/annurev-pharmtox-010716-104558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Tang XL, Hua Y, Guan Q, Yuan CH. Improved detection of deeply invasive candidiasis with DNA aptamers specific binding to (1–>3)-beta-D-glucans from Candida albicans. Eur J Clin Microbiol Infect Dis. 2016;35:587–595. doi: 10.1007/s10096-015-2574-8. [DOI] [PubMed] [Google Scholar]
  • 183.Mukherjee M, Bhatt P, H KM. Fluorescent competitive aptasensor for detection of aflatoxin B1. J Mol Recognit. 2017 doi: 10.1002/jmr.2650. [DOI] [PubMed] [Google Scholar]
  • 184.Wang SR, Min YQ, Wang JQ, Liu CX, Fu BS, Wu F. A highly conserved G-rich consensus sequence in Hepatitis C virus core gene represents a new anti-Hepatitis C target. Sci Adv. 2016;2:e1501535. doi: 10.1126/sciadv.1501535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Ren Y, Min YQ, Liu M, Chi L, Zhao P, Zhang XL. N-glycosylation-mutated HCV envelope glycoprotein complex enhances antigen-presenting activity and cellular and neutralizing antibody responses. Biochim Biophys Acta. 2016;1860:1764–1775. doi: 10.1016/j.bbagen.2015.08.007. [DOI] [PubMed] [Google Scholar]
  • 186.Zhao Y, Ren Y, Zhang X, Zhao P, Tao W, Zhong J. Ficolin-2 inhibits Hepatitis C Virus infection, whereas apolipoprotein E3 mediates viral immune escape. J Immunol. 2014;193:783–796. doi: 10.4049/jimmunol.1302563. [DOI] [PubMed] [Google Scholar]
  • 187.Lange MJ, Nguyen PDM, Callaway MK, Johnson MC, Burke DH. RNA-protein interactions govern antiviral specificity and encapsidation of broad spectrum anti-HIV reverse transcriptase aptamers. Nucleic Acids Res. 2017;45:6087–6097. doi: 10.1093/nar/gkx155. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Infection are provided here courtesy of Elsevier

RESOURCES