Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (265)

Search Parameters:
Keywords = SELEX

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 1401 KiB  
Review
Recent Developments in Aptamer-Based Sensors for Diagnostics
by Muhammad Sheraz, Xiao-Feng Sun, Yongke Wang, Jiayi Chen and Le Sun
Sensors 2024, 24(23), 7432; https://doi.org/10.3390/s24237432 - 21 Nov 2024
Abstract
Chronic and non-communicable diseases (NCDs) account for a large proportion of global disorders and mortality, posing significant burdens on healthcare systems. Early diagnosis and timely interference are critical for effective management and disease prevention. However, the traditional methods of diagnosis still suffer from [...] Read more.
Chronic and non-communicable diseases (NCDs) account for a large proportion of global disorders and mortality, posing significant burdens on healthcare systems. Early diagnosis and timely interference are critical for effective management and disease prevention. However, the traditional methods of diagnosis still suffer from high costs, time delays in processing, and infrastructure requirements that are usually unaffordable in resource-constrained settings. Aptamer-based biosensors have emerged as promising alternatives to offer enhanced specificity, stability, and cost-effectiveness for disease biomarker detection. The SELEX (Systematic Evolution of Ligands by Exponential Enrichment) methodology allows developing aptamers with high-affinity binding capabilities to a variety of targets, for instance proteins, cells, or even small molecules, hence rendering them suitable for NCD diagnosis. Aptasensors—recent developments in the electrochemical and optical dominion—offer much enhanced sensitivity, selectivity, and stability of detection across a diverse range of diseases from lung cancer and leukemia to diabetes and chronic respiratory disorders. This study provides a comprehensive review of progress in aptamer-based sensors, focusing on their role in point-of-care diagnostics and adaptability in a real-world environment with future directions in overcoming current limitations. Full article
(This article belongs to the Section Biosensors)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram of SELEX process showing the several stages taken by SELEX to produce aptamers. (1) Target incubation. (2) Dividing up. (3) Separation/elution. (4) PCR or RT-PCR amplification. (5) Cloning of chosen aptamer pool following the last SELEX phase [<a href="#B9-sensors-24-07432" class="html-bibr">9</a>].</p>
Full article ">Figure 2
<p>Aptasensors with electrochemical properties. Schematic illustration of the Fe(CN)<sub>6</sub><sup>4−/3−</sup> electrochemical aptasensor. (<b>a</b>) The structure of the aptamer included a hybridized form with complementary DNA that was fixed onto the gold surface. This aptamer was specifically designed to bind with the target, leading to a decrease in the number of aptamers present on the electrode surface when the target is detected. (<b>b</b>) This concept is illustrated in a schematic representation of the electrochemical aptasensor utilizing methylene blue (MB). (<b>c</b>) Another schematic diagram shows the Fc-based electrochemical aptasensor. When the target is present, the aptamer folds into a three-way junction that binds the target, which changes the electron transfer (eT) dynamics and results in a higher detected reduction peak. In the presence of the target, the aptamer takes on a constrained hairpin shape, and this change in conformation improves the efficiency of eT between the electrode surface and the ferrocene (Fc) probe [<a href="#B52-sensors-24-07432" class="html-bibr">52</a>,<a href="#B53-sensors-24-07432" class="html-bibr">53</a>].</p>
Full article ">Figure 3
<p>Diagrams showing optical aptasensors that use fluorescence: (<b>a</b>) show the most basic type of quenching aptamer beacon, in which the fluorescence is lowered due to the quencher and fluorophore being closer together because of target binding stabilizing the stem. (<b>b</b>) provides an example of the assembly aptamer beacon, in which oligomers assemble due to target binding, stabilizing the ternary complex. (<b>c</b>) displays the aptamer beacon after disassembly, when target binding causes antisense displacement to occur, increasing fluorescence [<a href="#B52-sensors-24-07432" class="html-bibr">52</a>].</p>
Full article ">Figure 4
<p>Schematic representations of optical aptasensors using AuNPs: (<b>a</b>) shows how target binding causes AuNPs to aggregate and release aptamers. (<b>b</b>) shows how target binding causes aptamers to release and AuNPs to disintegrate [<a href="#B52-sensors-24-07432" class="html-bibr">52</a>,<a href="#B66-sensors-24-07432" class="html-bibr">66</a>].</p>
Full article ">
15 pages, 2881 KiB  
Article
Development of a Graphene Oxide-Based Aptamer Nanoarray for Improved Neutralization and Protection Effects Against Ricin
by Huafei Li, Yanwen Ai, Yanjin Wu, Ruyu Fan, Yuan Tian, Shuangqun Chen, Wei Wan and Cong Wu
Pharmaceutics 2024, 16(11), 1455; https://doi.org/10.3390/pharmaceutics16111455 - 14 Nov 2024
Viewed by 431
Abstract
Background/Objectives: Ricin’s high toxicity and potential as a bioweapon underscore the need for effective antidotes. Monoclonal antibodies, though effective, are limited by complex production. This study aimed to develop a graphene oxide-based aptamer nanoarray (ARMAN) for improved neutralization and protection against ricin. [...] Read more.
Background/Objectives: Ricin’s high toxicity and potential as a bioweapon underscore the need for effective antidotes. Monoclonal antibodies, though effective, are limited by complex production. This study aimed to develop a graphene oxide-based aptamer nanoarray (ARMAN) for improved neutralization and protection against ricin. Methods: High-affinity aptamers targeting ricin’s RTA and RTB subunits were selected using SELEX technology and conjugated to graphene oxide (GO) via click chemistry. ARMAN’s characteristics, including morphology, stability, and biosecurity, were assessed. Its performance was evaluated in terms of affinity for ricin, neutralization capacity, and therapeutic effects in cellular assays and a mouse model of ricin poisoning. Results: ARMAN exhibited a uniform morphology with an average particle size of 217 nm and demonstrated significantly enhanced affinity for ricin compared to free aptamers. ARMAN showed rapid and effective neutralization ability, significantly increasing cell viability in BEAS-2B, GES-1, and HL7702 cell lines exposed to ricin. In vivo, ARMAN treatment led to a notable prolongation of survival in ricin-poisoned mice, highlighting its potential for both pre- and post-exposure treatment. These findings indicate that ARMAN not only neutralizes ricin effectively but also provides a therapeutic window for treatment. Conclusions: ARMAN’s superior binding affinity, serum stability, biocompatibility, and broad therapeutic efficacy make it a promising new antidote against ricin poisoning. This study’s findings represent significant progress in the development of rapid-response antidotes, with ARMAN offering a potential solution for both military and civilian emergency response scenarios. Full article
(This article belongs to the Special Issue New Nano-Systems for Imaging, Diagnostics, and Drug Delivery)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic diagram of the SELEX screening for aptamers targeting RTA and RTB. The red and blue symbols represent RTA and RTB specific aptamers, respectively.</p>
Full article ">Figure 2
<p>Generation and characterization of anti-RTA and anti-RTB aptamers. (<b>A</b>) Binding affinity of aptamer pools with RTA/B coupled microbeads after different round of SELEX by FCM. (<b>B</b>) Binding avidity of selected aptamers (RTA and RTB) with RTA/B coupled microbeads by FCM. (<b>C</b>) Purported secondary structure of the selected RTA and RTB aptamer.</p>
Full article ">Figure 3
<p>Fabrication and characterization of the ARMAN. (<b>A</b>) Schematic diagram of the construction process for the ARMAN, the red and blue symbols represent RTA and RTB specific aptamers, respectively. (<b>B</b>) Size distribution of GO and ARMAN determined by DLS. (<b>C</b>) AFM morphology of the ARMAN. Scale bar: 600 nm, thickness bar: 0 to 7 nm. (<b>D</b>) TEM morphology of the ARMAN. Scale bar: 500 nm. (<b>E</b>,<b>F</b>) FTIR spectra (<b>E</b>) and Zeta potential (<b>F</b>) of the ARMAN and counterparts.</p>
Full article ">Figure 4
<p>Favorable Serum Stability and Excellent Binding Affinity of the ARMAN. (<b>A</b>) Excellent in vitro serum stability of the ARMAN. DMEM containing 50% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) FBS was employed as an in vitro serum model, and the size distribution of the ARMAN was evaluated by DLS daily. (<b>B</b>) RT coupled magnetic beads were respectively incubated with the ARMAN and free aptamers at indicated concentrations from 25 nM to 800 nM, and the MFI was measured by FCM. The dissociation constant (Kd) was calculated based on the concentration MFI curve obtained using IBM SPSS Software (Version 29.0.2). (<b>C</b>) Binding off-rate of the ARMAN and parental aptamers from RT coupled magnetic beads. Beads were incubated with fluorescence-labeled aptamer or ARMAN at a concentration of 400 nM. Following incubation, the beads were washed at various time points, and the MFI was assessed by FCM. The proportion of aptamers remaining on the surface of the magnetic beads was subsequently calculated and compared. Data are expressed as means ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>Favorable biocompatibility of the ARMAN and free aptamer counterparts. (<b>A</b>) Hemolysis quantification of RBC incubated with the ARMAN and free aptamers. (<b>B</b>–<b>H</b>) Blood routine and biochemical indexes analysis of mice in plasma samples collected from healthy mice 24 h after tail vein injection of the ARMAN and its counterparts. Data are expressed as means ± SD (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>Exceptional protective effects of the ARMAN in vitro. (<b>A</b>–<b>C</b>) CCK-8 assays were conducted to measure the viability of cells exposed to RT and various antidotes. (<b>D</b>–<b>F</b>) Cells were incubated with 4 ng/mL of Ricin toxin, and 400 nM of free aptamers and ARMAN were used for detoxification. The cell viability ratios of each treatment group to the control group were calculated and compared. Data are expressed as means ± SD (<span class="html-italic">n</span> = 3), ** <span class="html-italic">p</span> &lt; 0.01; ns: not significant.</p>
Full article ">Figure 7
<p>Outstanding protective effects of the ARMAN in vivo. (<b>A</b>) For the detoxification experiment, mice were administered tail vein injections of the ARMAN and its counterparts with an equivalent aptamer amount at 15 min, 1 h, and 2 h post RT exposure. (<b>B</b>) For the prevention–detoxification experiment, the ARMAN and its counterparts were injected 15 min before and 1 and 2 h after Ricin administration. The mice were closely monitored daily for vital signs until their natural death within 14 days.</p>
Full article ">
17 pages, 3538 KiB  
Article
Novel DNA Aptamers to Dickkopf-1 Protein and Their Application in Colorimetric Sandwich Assays for Target Detection in Patients with Axial Spondyloarthritis
by Elizaveta A. Shatunova, Anastasia S. Rychkova, Mariya I. Meschaninova, Marsel R. Kabilov, Alexey E. Tupikin, Yuliya D. Kurochkina, Maksim A. Korolev and Mariya A. Vorobyeva
Int. J. Mol. Sci. 2024, 25(22), 12214; https://doi.org/10.3390/ijms252212214 - 14 Nov 2024
Viewed by 225
Abstract
Chronic immunoinflammatory rheumatic diseases, such as axial spondyloarthritis (AxSpA), are accompanied by a dysregulation of bone remodeling. Among potential biomarkers of bone metabolism, the Wnt pathway antagonist, Dickkopf-1 (DKK-1), is of particular interest because of its potential to reflect a shift towards joint [...] Read more.
Chronic immunoinflammatory rheumatic diseases, such as axial spondyloarthritis (AxSpA), are accompanied by a dysregulation of bone remodeling. Among potential biomarkers of bone metabolism, the Wnt pathway antagonist, Dickkopf-1 (DKK-1), is of particular interest because of its potential to reflect a shift towards joint ossification or osteoporosis, but its diagnostic value needs validation. There is still a lack of stable and efficient methods of measuring serum DKK-1 levels suitable for longitude studies. The use of aptamer-based diagnostic assays could be very promising for this purpose. We generated novel anti-DKK-1 DNA aptamers from a combinatorial library with a pre-defined sequence pattern in the randomized region. This approach showed high efficacy, as only four SELEX rounds of selection produced high-affinity aptamers with dissociation constants ranging from 1.3 to 3.7 nM. A family of their truncated versions was also developed by rational design. Novel DNA aptamers functioned as capture components in a microplate ELISA-like assay with HRP-conjugated anti-DKK-1 antibody as a reporter component. We succeeded in revealing the aptamer/aptamer sandwich pairs that provided an aptamer-only sandwich colorimetric assay. The aptamer/antibody colorimetric test systems were also examined in the analyses of blood serum from AxSpA patients and shown sufficient workability. However, in a number of cases we registered significant differences between assays based on TD10 and DK4 aptamers and made some suggestions about the origin of this effect. Full article
Show Figures

Figure 1

Figure 1
<p>An assessment of library enrichment evaluation after the 4th round of selection: Non-denaturing PAGE analysis of Cy5-labeled DNA–protein complexes (<b>A</b>) and corresponding binding curve generated with a bimolecular binding equation in GraphPad Prism 8.0.1 software (<b>B</b>).</p>
Full article ">Figure 2
<p>The most probable secondary structures of aptamers DK1–DK5 were obtained with the mfold web server [<a href="#B27-ijms-25-12214" class="html-bibr">27</a>]. Transversions in the pyrimidine–purine alternation pattern are shown in red boxes.</p>
Full article ">Figure 3
<p>ELASA analysis of aptamer binding with DKK1. Microplate wells were coated with 2 μg/mL DKK-1; the concentration of aptamers varied from 3.12 nM to 100 nM. Each data point represents the average value of two independent experiments.</p>
Full article ">Figure 4
<p>Schematic representation of rational truncation for aptamers DK1 (<b>A</b>), DK2 (<b>B</b>), and DK4 (<b>C</b>). AT pairs are shown by black lines, GC pairs by red lines.</p>
Full article ">Figure 5
<p>ELASA analysis of truncated DNA aptamer binding with DKK1. Microplate’s wells were coated with 2 μg/mL DKK-1; the concentration of aptamers varied from 3.12 nM to 100 nM. Each data point represents the average value of two independent experiments.</p>
Full article ">Figure 6
<p>Schematic representation of colorimetric sandwich systems for DKK-1 detection, based on aptamer/antibody (<b>A</b>), antibody/aptamer (<b>B</b>), or aptamer/aptamer (<b>C</b>) pairs.</p>
Full article ">Figure 7
<p>Dependencies of optical density from DKK-1 concentration for aptamer/antibody sandwich systems; the GraphPad Prism 8.0.1 software package was used to plot the 4PL curve. Each point represents the average value of two independent experiments.</p>
Full article ">Figure 8
<p>Dependencies of optical density from DKK-1 concentration for aptamer/aptamer sandwich pairs DK1/DK4 (<b>A</b>), DK1_48t/DK4_41t (<b>B</b>), DK1_48t/DK4_50t (<b>C</b>), DK4/TD10 (<b>D</b>), and DK4_41t/TD10 (<b>E</b>). The GraphPad Prism 8.0.1 software package was used to plot the linear dependency of OD450 from DKK-1 concentration. Each point represents the average value of two independent experiments.</p>
Full article ">Figure 9
<p>Results of the DK4-based (red bars), TD10-based (gray bars), and ELISA (black bars) analyses of the DKK-1 levels in blood serums diluted by PBS (1:10). The values of DKK-1 concentration in the samples were calculated with the use of the online service <a href="https://www.myassays.com/four-parameter-logistic-curve.assay" target="_blank">https://www.myassays.com/four-parameter-logistic-curve.assay</a> (accessed on 7 November 2024). Each point represents the average value of two independent experiments.</p>
Full article ">
16 pages, 7688 KiB  
Article
Enhanced Detection of Vibrio harveyi Using a Dual-Composite DNAzyme-Based Biosensor
by Siying Li, Shuai Zhang, Weihong Jiang, Yuying Wang, Mingwang Liu, Mingsheng Lyu and Shujun Wang
Biosensors 2024, 14(11), 548; https://doi.org/10.3390/bios14110548 - 13 Nov 2024
Viewed by 451
Abstract
Vibrio harveyi is a serious bacterial pathogen which can infect a wide range of marine organisms, such as marine fish, invertebrates, and shrimp, in aquaculture, causing severe losses. In addition, V. harveyi can be transmitted through food and water, infecting humans and posing [...] Read more.
Vibrio harveyi is a serious bacterial pathogen which can infect a wide range of marine organisms, such as marine fish, invertebrates, and shrimp, in aquaculture, causing severe losses. In addition, V. harveyi can be transmitted through food and water, infecting humans and posing a serious threat to public safety. Therefore, rapid and accurate detection of this pathogen is key for the prevention and control of related diseases. In this study, nine rounds of in vitro screening were conducted with Systematic Evolution of Ligands by Exponential Enrichment (SELEX) technology using unmodified DNA libraries, targeting the crude extracellular matrix (CEM) of V. harveyi. Two DNAzymes, named DVh1 and DVh3, with high activity and specificity were obtained. Furthermore, a fluorescent biosensor with dual DNAzymes was constructed which exhibited improved detection efficiency. The sensor showed a good fluorescence response to multiple aquatic products (i.e., fish, shrimp, and shellfish) infected with V. harveyi, with a detection limit below 11 CFU/mL. The fluorescence signal was observed within 30 min of reaction after target addition. This simple, inexpensive, highly effective, and easy to operate DNAzymes biosensor can be used for field detection of V. harveyi. Full article
(This article belongs to the Section Optical and Photonic Biosensors)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flowchart of DNAzyme screening. The library contained 40 random nucleotides and was screened for nine rounds. Negative selection was carried out during the 5th and 7th rounds of screening, while positive selections were conducted in the other rounds. Biotin was labeled at the 5′ end. The target molecule was the CEM of bacteria.</p>
Full article ">Figure 2
<p>Activity of candidate DNAzymes: (<b>a</b>) fluorescence intensity (DVh1–6 reacted with CEM-Vh); and (<b>b</b>) results of candidate 15% dPAGE (DVh1–6 reacted with CEM-Vh, Blank1–6 mean DVh1–6 reacted without CEM-Vh).</p>
Full article ">Figure 3
<p>Differences between the fluorescence intensity signals of single and dual DNAzymes. (Blank means without adding CEM-Vh, CEM-Vh means adding CEM-Vh. Different letters indicate statistically significant differences (<span class="html-italic">p</span> &lt; 0.05), while identical letters indicate insignificant differences (<span class="html-italic">p</span> &gt; 0.05)).</p>
Full article ">Figure 4
<p>Optimization of reaction conditions: (<b>a</b>) pH optimization; (<b>b</b>) influence of various divalent metal ions on the cleavage activity of DVh<sub>3+1</sub> (different letters indicate statistically significant differences (<span class="html-italic">p</span> &lt; 0.05), while identical letters indicate insignificant differences (<span class="html-italic">p</span> &gt; 0.05)); and (<b>c</b>) optimization of the concentrations of Na<sup>+</sup> and Mg<sup>2+</sup>. Buffer/EDTA reaction contained 300 mM EDTA in 2× selection buffer. The bars and the dots represent mean ± SD.</p>
Full article ">Figure 5
<p>Specificity of DVh<sub>3+1</sub>: (<b>a</b>) fluorescence intensity of DVh<sub>3+1</sub> in presence of CEM of various bacteria; (<b>b</b>) specificity of DVh<sub>3+1</sub> analyzed by 15% dPAGE (Blank: reaction system without CEM-Vh).</p>
Full article ">Figure 6
<p>Sensitivity of DVh<sub>3+1</sub>: (<b>a</b>) fluorescence intensity signals of different concentrations of <span class="html-italic">V. harveyi</span> (Blank: normal saline) and the calibration curves constructed using the fluorescence signals corresponding to 4.7 × 10<sup>1</sup>, 4.7 × 10<sup>2</sup>, and 4.7 × 10<sup>3</sup> CFU/mL of <span class="html-italic">V. harveyi</span>; and (<b>b</b>) gel cleavage assay at different concentrations of <span class="html-italic">V. harveyi</span> (Blank: reaction system without CEM-Vh).</p>
Full article ">Figure 7
<p>Identification of the target of DVh<sub>3+1</sub>: (<b>a</b>) fluorescence intensity of untreated and protease-treated CEM-Vh cleaved by DVh<sub>3+1</sub>; (<b>b</b>) 15% dPAGE analysis of the cleavage activity of DVh<sub>3+1</sub> against CEM-Vh treated with various proteases; (<b>c</b>) fluorescence intensity of the reactions between DVh<sub>3+1</sub> and CEM-Vh with different molecular weights; (<b>d</b>) 15% dPAGE analysis of the cleavage activity of DVh<sub>3+1</sub> against CEM-Vh with different molecular weights. (Blank: reaction system without CEM-Vh).</p>
Full article ">Figure 8
<p>Effect of four different RNases on the cleavage activity of DVh<sub>3+1</sub> (Blank: reaction system without CEM-Vh; Different letters indicate statistically significant differences (<span class="html-italic">p</span> &lt; 0.05), while identical letters indicate insignificant differences (<span class="html-italic">p</span> &gt; 0.05)).</p>
Full article ">Figure 9
<p>(<b>a</b>) Optimization of the concentration of DVh<sub>3+1</sub>-S in the dual DNAzyme sensor, with corresponding pictures of fluorescence signal shown at the top. (<b>b</b>) Analysis of significant differences between the fluorescence values of different concentrations of DVh<sub>3+1</sub>-S at different reaction times. Different letters indicate statistically significant differences (<span class="html-italic">p</span> &lt; 0.05), while the same letters indicate insignificant differences (<span class="html-italic">p</span> &gt; 0.05). The bar represents the mean ± SD.</p>
Full article ">Figure 10
<p>Comparisons of the aquatic products infected by <span class="html-italic">V. harveyi</span>: (<b>a</b>) <span class="html-italic">P. vannamei</span> before infection, (<b>b</b>) <span class="html-italic">P. vannamei</span> after infection 24 h, (<b>c</b>) whelk before infection, (<b>d</b>) Whelk after infection 60 h, (<b>e</b>) <span class="html-italic">C. formosana</span> before infection, (<b>f</b>) <span class="html-italic">C. formosana</span> after infection 48 h, (<b>g</b>) <span class="html-italic">Epinephelus</span> before infection and (<b>h</b>) <span class="html-italic">Epinephelus</span> after infection 50 h.</p>
Full article ">Figure 11
<p><span class="html-italic">V. harveyi</span> detection in the actual samples by DVh<sub>3+1</sub> sensor: (<b>a</b>) fluorescence intensities of blank and four actual samples (<span class="html-italic">C. formosana</span>, <span class="html-italic">Epinephelus</span>, <span class="html-italic">P. vannamei</span>, and whelk; Blank: uninfected animal samples); (<b>b</b>) fluorescence intensities of diluted whelk samples, with calibration curves plotted using the fluorescence values of <span class="html-italic">V. harveyi</span> at concentrations of 1.02 × 10<sup>1</sup>, 1.02 × 10<sup>2</sup>, and 1.02 × 10<sup>3</sup> CFU/mL (Different letters indicate statistically significant differences (<span class="html-italic">p</span> &lt; 0.05)).</p>
Full article ">
14 pages, 2344 KiB  
Article
Matrix Background Screening of an ssDNA Aptamer and Its Identification Against Lactopontin
by Chao Zhu, Ziru Feng, Mengmeng Yan, Hongxia Du, Tengfei Li and Jiangsheng Mao
Int. J. Mol. Sci. 2024, 25(21), 11832; https://doi.org/10.3390/ijms252111832 - 4 Nov 2024
Viewed by 543
Abstract
Lactopontin (LPN) is a highly phosphorylated O-glycosylated acidic protein closely associated with infant gut, brain, and immune development, and its recognition is urgent due to its rising application in fortified dairy products and infant formula. In this study, an ssDNA aptamer against LPN [...] Read more.
Lactopontin (LPN) is a highly phosphorylated O-glycosylated acidic protein closely associated with infant gut, brain, and immune development, and its recognition is urgent due to its rising application in fortified dairy products and infant formula. In this study, an ssDNA aptamer against LPN was obtained, among which two kinds of matrix-background-assisted systematic evolution of ligands via exponential enrichment (SELEX) approaches were performed and compared. The direct approach was to utilize the sample matrix as the mixing-incubation background between the ssDNA library and LPN that can theoretically increase screening pressure and simulate practical application scenarios. The indirect approach was to utilize a PBS buffer as a screening background and to include counter-screening steps that adopt the “sample matrix” as a whole as the counter-screening target. Their screening evolutions were monitored through qPCR assays from sequence diversity convergences of each sub-library based on the change in the proportion of hetero- and homo-duplexes from the dissociation curve and melting temperature, which were also verified from the sequence statistics of high-throughput sequencing. The common sequence of Seq.I1II3 from the two approaches was finally fished out as the aptamer through multiple analyses of combining the sequence frequency, secondary structures, homology, and binding assessments, which was demonstrated good specificity and low-nanomolar affinity by qPCR assay (KD, 5.9 nM). In addition, molecular docking and a dynamics simulation were performed for their binding site prediction and affinity confirmation. This study provides a potential identifying element and a basis for accelerating the development of methods for LPN detection in dairy products. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the direct (<b>A</b>) and indirect (<b>B</b>) matrix background SELEX approaches.</p>
Full article ">Figure 2
<p>The dissociation curve analysis for each library to monitor the screening evolution: ten rounds (<b>A</b>) and the clear curves’ changes (<b>C</b>) in the direct approach; eight rounds (<b>B</b>) and the clear curves’ changes (<b>D</b>) in the indirect approach; their separate AGE results (<b>E</b>,<b>F</b>) of the PCR products after linking the connectors before HTS changed from 80 bp (bands 1 and 3) to the position of 200 bp (bands 2 and 4).</p>
Full article ">Figure 3
<p>(<b>A</b>): Evolutionary tree of the top 10 sequences of the direct approach (top) and indirect approach (bottom) via MEGA-11. (<b>B</b>): Their sequence logos of conserved bases through a Clustal X analysis of the direct approach (Top) and indirect approach (bottom). (<b>C</b>): The motif analysis of the selected nine candidate sequences through MEME Suite.</p>
Full article ">Figure 4
<p>(<b>A</b>): The ssDNA standard solutions through qPCR assay between ssDNA concentration and Cq values. (<b>B</b>): The preliminary affinity evaluation of the nine selected candidate sequences. (<b>C</b>): The affinity of Seq.I1II3 with seven concentration (Conc.) levels (0, 5, 10, 20, 50, 100, and 200 nM) based on the non-linear fitting method. (<b>D</b>): The specificity assessment of Seq.I1II3 (**** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 5
<p>Binding mode analysis between the aptamer and LPN. The accompanying figure delineates the specific binding site with amino acid residues represented as blue stick models and nucleotide residues as red stick models.</p>
Full article ">
15 pages, 3519 KiB  
Article
A Double-Stranded Aptamer for Highly Sensitive Fluorescent Detection of Glutathione S-Transferases
by Wei Cui, Suping Li, Jiahao Zeng, Chen Li, Zhaofeng Li, Xiaohong Wen, Suxia Bao, Yang Mei, Xiangxian Meng and Qiuping Guo
Biosensors 2024, 14(10), 476; https://doi.org/10.3390/bios14100476 - 3 Oct 2024
Viewed by 933
Abstract
Aptamer-based biosensors have been widely constructed and applied to detect diverse targets. Glutathione S-transferase (GST), a pivotal phase II metabolic enzyme, plays a critical role in biotransformation in vivo, and aberrant GST expression is associated with various health risks. Herein, aptamers targeting GST [...] Read more.
Aptamer-based biosensors have been widely constructed and applied to detect diverse targets. Glutathione S-transferase (GST), a pivotal phase II metabolic enzyme, plays a critical role in biotransformation in vivo, and aberrant GST expression is associated with various health risks. Herein, aptamers targeting GST were systematically selected from a randomized single-stranded DNA (ssDNA) library of 79 nucleotides (nt) using a biotinylated GST-immobilized streptavidin agarose (SA) bead SELEX technology. Following rigorous screening across eight rounds, four aptamers with strikingly similar secondary structures emerged. Among these, Seq3 exhibited the highest affinity towards GST and was selected for further optimization. A semi-rational post-SELEX truncation strategy was then employed based on base composition analysis, secondary structure analysis and affinity assessment. This strategy enabled the systematic removal of redundant nucleotides in Seq3 without compromising its affinity, ultimately yielding a truncated aptamer, Seq3-3, which retains its specificity with a compact 39nt length. Building upon Seq3-3, a double-stranded fluorescent aptamer probe was ingeniously designed for the in vitro detection of GST. The detection mechanism hinges on the competitive displacement of the complementary chain from the probe, mediated by the target protein, leading to the separation of the antisense oligonucleotide from the double-stranded complex. This process triggers the restoration of the fluorescence signal, enabling sensitive detection, and the probe exhibits excellent response within a linear range of GST activity ranging from 0 to 1500 U/L. The results show that not only an efficient strategy for screening robust and practicable aptamers but also an ultrahighly sensitive detection platform for GST was established. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic diagram of protein-SELEX screening process.</p>
Full article ">Figure 2
<p>Confocal imaging fluorescence intensity verified the enrichment degree of the library.</p>
Full article ">Figure 3
<p>Determination and specificity of sequence dissociation equilibrium constant: (<b>A</b>) the saturation curves and Kd values of Seq3, Seq4, Seq7, and Seq20; and (<b>B</b>) investigation of sequence specificity.</p>
Full article ">Figure 4
<p>Effect of conditional optimization on the binding ability of Seq3 sequences. (<b>A</b>) The binding ability of temperature to Seq3 sequence was investigated. (<b>B</b>) Sequence optimization of aptamer Seq3, showing its secondary structure as predicted using the Mfold software (version 2.1).</p>
Full article ">Figure 5
<p>Investigation of sequence affinity after optimization. (<b>A</b>) Confocal imaging was used to investigate the sequence binding ability after optimization. Scale: 100 μm. (<b>B</b>) The modified sequence design method of Seq3-3 truncated the front and rear primers at the same time and still maintained a high fluorescence intensity level. (<b>C</b>) The effect of temperature on the combination of optimized sequences was investigated. Red mark: Seq3-3 had the best activity in combination with GST, so it was selected to continue the follow-up experiment.</p>
Full article ">Figure 6
<p>Design of double-strand fluorescent aptamer probe: (<b>A</b>) schematic diagram; (<b>B</b>) screening of cDNA; and (<b>C</b>) aptamer/cDNA5 ratio optimization.</p>
Full article ">Figure 7
<p>Study on the activity of double-strand fluorescent aptamer probe for GST: (<b>A</b>) detection of GST on agarose microbeads; (<b>B</b>) detection of GST in eluents; (<b>C</b>) detection of GST on elution remaining microbeads; (<b>D</b>) fluorescence spectrum of GST quantitative detection; and (<b>E</b>) the standard curve in D.</p>
Full article ">Figure 8
<p>Study on the specificity of double-strand fluorescent aptamer probe for GST: (<b>A</b>) effect of aptamer on enzymatic reaction of GST; and (<b>B</b>) investigation of specificity of double-strand fluorescent probe.</p>
Full article ">
19 pages, 3040 KiB  
Article
Identification of Podoplanin Aptamers by SELEX for Protein Detection and Inhibition of Platelet Aggregation Stimulated by C-Type Lectin-like Receptor 2
by Hui-Ju Tsai, Kai-Wen Cheng, Jou-Chen Li, Tsai-Xiang Ruan, Ting-Hsin Chang, Jin-Ru Wang and Ching-Ping Tseng
Biosensors 2024, 14(10), 464; https://doi.org/10.3390/bios14100464 - 27 Sep 2024
Viewed by 859
Abstract
Tumor cell-induced platelet aggregation (TCIPA) is a mechanism for the protection of tumor cells in the bloodstream and the promotion of tumor progression and metastases. The platelet C-type lectin-like receptor 2 (CLEC-2) can bind podoplanin (PDPN) on a cancer cell surface to facilitate [...] Read more.
Tumor cell-induced platelet aggregation (TCIPA) is a mechanism for the protection of tumor cells in the bloodstream and the promotion of tumor progression and metastases. The platelet C-type lectin-like receptor 2 (CLEC-2) can bind podoplanin (PDPN) on a cancer cell surface to facilitate TCIPA. Selective blockage of PDPN-mediated platelet–tumor cell interaction is a plausible strategy for inhibiting metastases. In this study, we aimed to screen for aptamers, which are the single-stranded DNA oligonucleotides that form a specific three-dimensional structure, bind to specific molecular targets with high affinity and specificity, bind to PDPN, and interfere with PDPN/CLEC-2 interactions. The systematic evolution of ligands by exponential enrichment (SELEX) was employed to enrich aptamers that recognize PDPN. The initial characterization of ssDNA pools enriched by SELEX revealed a PDPN aptamer designated as A1 displaying parallel-type G-quadruplexes and long stem-and-loop structures and binding PDPN with a material with a dissociation constant (Kd) of 1.3 ± 1.2 nM. The A1 aptamer recognized both the native and denatured form of PDPN. Notably, the A1 aptamer was able to quantitatively detect PDPN proteins in Western blot analysis. The A1 aptamer could interfere with the interaction between PDPN and CLEC-2 and inhibit PDPN-induced platelet aggregation in a concentration-dependent manner. These findings indicated that the A1 aptamer is a candidate for the development of biosensors in detecting the levels of PDPN expression. The action by A1 aptamer could result in the prevention of tumor cell metastases, and if so, could become an effective pharmacological agent in treating cancer patients. Full article
(This article belongs to the Special Issue Design and Application of Novel Nucleic Acid Probe)
Show Figures

Figure 1

Figure 1
<p>SELEX for the screening of the PDPN aptamer. (<b>A</b>) Schematic representation of the selection process of SELEX employed to identify PDPN aptamer. (<b>B</b>) The binding of PDPN (100 nM) to the initial aptamer library (original) or the ssDNA pool obtained from the ninth round of SELEX screening (Ninth round) was performed using the BLI assay. Representative binding intensity during the association and dissociation of PDPN to the initial aptamer library and the ssDNA pool coated on the chip are shown.</p>
Full article ">Figure 2
<p>The characterization of putative PDPN aptamers. (<b>A</b>) The alignment of six representative aptamer sequences by using Bioedit software. (<b>B</b>) Aptamers were coated on the BLI chip and incubated with the indicated concentrations of PDPN. The dissociation constant K<sub>d</sub> was calculated, as shown in <a href="#biosensors-14-00464-t001" class="html-table">Table 1</a>. (<b>C</b>) The dot blot analysis for binding between PDPN and the six putative PDPN aptamers. PDPN (50 nM) was spotted on the nitrocellulose membrane followed by subsequent incubation with the indicated biotin-labeled aptamers (80 nM) and the streptavidin-conjugated HRP. The binding of the anti-PDPN antibody (NZ-1) to the PDPN spotted on the nitrocellulose membrane was used as a positive control. (<b>D</b>) Statistical analysis for the binding between PDPN and the six putative PDPN aptamers in dot blot analysis, as shown in panel (<b>C</b>). The binding intensity of A1 to PDPN was arbitrarily set as 1. The data represent the mean ± SEM of 2 to 3 independent experiments. *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.001; ***, <span class="html-italic">p</span> &lt; 0.0001; ns, no significance.</p>
Full article ">Figure 3
<p>Schematic illustrations of the steric structures of the putative PDPN aptamers. (<b>A</b>,<b>B</b>). The sequences of the indicated PDPN aptamers were subjected to structural analyses using the structure prediction softwares NUPACK 4.0 (panel (<b>A</b>)) and mFold (panel (<b>B</b>)). Stem and loop structures were formed.</p>
Full article ">Figure 4
<p>CD spectra of the six putative PDPN aptamers. The aptamers were denatured at 95 °C for 5 min and cooled on ice for at least 5 min before analysis. The CD spectra were measured using a J-815 CD Spectrometer (JASCO Corporation, Hachioji, Japan). The six aptamers displayed distinctive G-quadruplex patterns.</p>
Full article ">Figure 5
<p>The binding specificity of A1 aptamer to PDPN. (<b>A</b>) PDPN-His-Fc or IgG-Fc (50 nM) was coated on the BLI chip and incubated with the A1 aptamer for BLI assay. The association and dissociation curves for the binding of the A1 aptamer with the recombinant PDPN and IgG-Fc are shown. (<b>B</b>) Western blot analysis was performed for the lysates from HEK293T cells transfected with the indicated expression plasmid. Protein expression was detected by using the anti-CLEC-2 antibody (left panel) or the anti-His-Tag antibody (right panel). (<b>C</b>) Flow cytometry analysis of CLEC-2 expression on the cell surface. The HEK293T cells were transfected with the indicated plasmids encoding CLEC-2 protein with or without the His-Tag sequence. At 48 h after transfection, the cells were subjected to flow cytometry analysis using the primary mouse anti-human CLEC-2 antibody and the secondary FITC-conjugated goat anti-mouse antibody. Representative histograms are shown. (<b>D</b>) HEK293T cells were transfected with the plasmids encoding the indicated proteins. At 48 h after transfection, the cells were incubated with the FAM-conjugated A1 aptamer (1 μM). The percentages of cells with A1 binding were analyzed by flow cytometry. The data represent the mean ± SEM of three independent experiments. (<b>E</b>) The indicated cell lines were incubated with different concentrations of the FAM-conjugated A1 aptamer. Flow cytometry was performed to determine the levels of A1 binding on the cell surface. The data represent the mean ± SEM of three independent experiments. ns, no significance; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>The quantitative detection of PDPN by the aptamer A1. (<b>A</b>) The indicated amounts of PDPN protein were subjected to Western blot analysis. The biotin-labeled aptamer A1 and the streptavidin-conjugated HRP were used to detect PDPN protein by ECL. (<b>B</b>) The linear correlations of the signal intensities and the amounts of PDPN present in the samples were performed. The data represent the mean ± SEM of four independent experiments. The linear correlation equation and the R<sup>2</sup> value are shown.</p>
Full article ">Figure 7
<p>A1 aptamer interferes with PDPN/CLEC-2 interactions. (<b>A</b>) The recombinant PDPN protein was spotted on the nitrocellulose membrane and incubated with the indicated concentrations of the CLEC-2 protein. The binding of CLEC-2 to the spotted PDPN was detected using the anti-CLEC-2 antibody followed by the secondary antibody. ECL was performed to detect the signal. (<b>B</b>) PDPN was spotted on the nitrocellulose membrane. The A1 aptamer (A1), A1498 aptamer (A1498), or anti-PDPN antibody NZ-1 (PC) and the CLEC-2 proteins were incubated with the membrane. The amount of CLEC-2 binding to PDPN was determined by incubating the membrane with the anti-CLEC-2 antibody followed by the secondary antibody. ECL was performed to detect the signal. Quantification was performed using ImageJ. The amount of CLEC-2 binding to PDPN without aptamer or antibody (NC group) was arbitrarily set to 100%. The data represent the mean ± SEM of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ns, no significance.</p>
Full article ">Figure 8
<p>Aptamer A1 inhibits PDPN-induced platelet aggregation. (<b>A</b>,<b>B</b>) PDPN-induced platelet aggregation was performed using the 96-well plate method. The kinetics of platelet aggregation in the presence or absence of the indicated aptamers or anti-PDPN antibody NZ-1 were determined by the continuous measurement of light absorbance at OD595 nm using the SpectraMax Plus 384 Microplate Reader (Marshall Scientific, Hampton, NH, USA). Representative platelet aggregation traces for the indicated treatments are shown (panel (<b>A</b>)). The degree of aggregation at the end of the assay was determined. The data represent the mean ± SEM of three independent experiments (panel (<b>B</b>)). *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ns, no significance.</p>
Full article ">
18 pages, 22028 KiB  
Article
A Spatiotemporal Transcriptome Reveals Stalk Development in Pearl Millet
by Fei Mao, Lin Luo, Nana Ma, Qi Qu, Hao Chen, Chao Yi, Mengxue Cao, Ensi Shao, Hui Lin, Zhanxi Lin, Fangjie Zhu, Guodong Lu and Dongmei Lin
Int. J. Mol. Sci. 2024, 25(18), 9798; https://doi.org/10.3390/ijms25189798 - 10 Sep 2024
Viewed by 725
Abstract
Pearl millet is a major cereal crop that feeds more than 90 million people worldwide in arid and semi-arid regions. The stalk phenotypes of Poaceous grasses are critical for their productivity and stress tolerance; however, the molecular mechanisms governing stalk development in pearl [...] Read more.
Pearl millet is a major cereal crop that feeds more than 90 million people worldwide in arid and semi-arid regions. The stalk phenotypes of Poaceous grasses are critical for their productivity and stress tolerance; however, the molecular mechanisms governing stalk development in pearl millet remain to be deciphered. In this study, we spatiotemporally measured 19 transcriptomes for stalk internodes of four different early developmental stages. Data analysis of the transcriptomes defined four developmental zones on the stalks and identified 12 specific gene sets with specific expression patterns across the zones. Using weighted gene co-expression network analysis (WGCNA), we found that two co-expression modules together with candidate genes were involved in stalk elongation and the thickening of pearl millet. Among the elongation-related candidate genes, we established by SELEX that a MYB-family transcription factor PMF7G02448 can bind to the promoters of three cell wall synthases genes (CesAs). In summary, these findings provide insights into stalk development and offer potential targets for future genetic improvement in pearl millet. Full article
(This article belongs to the Collection Advances in Molecular Plant Sciences)
Show Figures

Figure 1

Figure 1
<p>Transcriptomes of internodes reveal four developmental zones of stalks. (<b>a</b>) Stalk of pearl millet at four developmental stages. (FS: first stage; SS: second stage; TS: third stage; LS: last stage). Scale: red bars, 1 cm. (<b>b</b>) Cluster dendrogram showing the similarities and differences among internode samples based on transcriptomic data. This analysis reveals distinct clusters corresponding to different developmental zones (Zone I–IV), each representing a distinct developmental stage based on gene expression profiles. (<b>c</b>) Principal component analysis (PCA) plot indicating the first two principal components (PC1 and PC2), which account for 43.47% of the total variance among samples. The plot classifies the samples into Zone I–IV. Colors represent different developmental zones: Zone I, Zone II, Zone III, Zone IV.</p>
Full article ">Figure 2
<p>Spatiotemporal pattern of gene expressions and functional enrichment analysis. (<b>a</b>) Expression patterns of 19,887 high-variation genes (HVGs) in pearl millet internode samples, classified into 12 clusters (C1–C12) using the fuzzy c-means clustering algorithm. Twelve clusters (C1–C12) were defined. Internodes belonging to different developmental zones are indicated with the background colors. Colored lines represent the gene expression profiles across different internode samples within each cluster. (<b>b</b>) Enrichment of functional categories of the twelve fuzzy c-means clusters.</p>
Full article ">Figure 3
<p>Expression patterns of HVTFs. (<b>a</b>) Number (bar plot) and proportion (colored square) of HVTFs in each TF family. For each TF family, the ratio equals the number of HVTFs divided by the number of TFs. (<b>b</b>) The expression of all HVTFs in the 19 stalk internodes. The red line indicates that LOG<sub>2</sub>(TPM+1) equals four. (<b>c</b>) Clustering heatmap shows six groups of expression patterns of all HVTFs. (<b>d</b>) The proportion of HVTFs in each TF family in the 6 groups defined in (<b>c</b>).</p>
Full article ">Figure 4
<p>Co-expression network analysis identifies gene modules related to stalk length and diameter. (<b>a</b>) Gene modules and their correlation with stalk length and diameter. The number of genes, the correlation coefficient, and the <span class="html-italic">p</span> value of the correlation (in brackets) are indicated for each module. The correlation coefficient between the module and trait is shown in red for positive correlations (ranging from 0 to 1) and blue for negative correlations (ranging from −1 to 0). (<b>b</b>) Scatter plot for genes in the pink module, illustrating the correlation between their module membership and their effect significance on stalk length. (<b>c</b>) Scatter plot for genes in the purple module, illustrating the correlation between their module membership and their effect significance on stalk diameter. (<b>d</b>,<b>e</b>) The expression profile of all genes in the pink module (<b>d</b>) and the purple module (<b>e</b>). The color scales represents the Z score of expression level. (<b>f</b>,<b>g</b>) Enrichment of functional categories for genes in the pink module (<b>f</b>) and the purple module (<b>g</b>).</p>
Full article ">Figure 5
<p>Candidate genes regulating stalk length of pearl millet. (<b>a</b>) Phylogenetic tree of CesA family proteins in Arabidopsis (with “At” prefix), rice (with “LOC_Os” prefix), and pearl millet (with “PMF” prefix). (<b>b</b>) Venn diagram showing that 4 <span class="html-italic">PmCesA</span> genes belong to both the pink module and the <span class="html-italic">CesA</span> family genes. (<b>c</b>) The expression pattern of the four <span class="html-italic">PmCesA</span> genes; note that three of them are expressed more consistently with that of the pink module (<a href="#ijms-25-09798-f004" class="html-fig">Figure 4</a>d). (<b>d</b>) Contents of cellulose, hemicellulose, and lignin in the internodes of pearl millet stalks. Error bars represent ± SD (n = 3). (<b>e</b>) The correlation between expressions of <span class="html-italic">PmCesA</span> and TFs in the internodes of pearl millet stalks. Names of highly correlated TFs are in red. (<b>f</b>) Co-expression network showing genes of top-100 highest connectivity in the pink module. Yellow circles represent <span class="html-italic">PmCesA</span>, red circles represent the highly correlated TFs in (<b>e</b>), and other genes are represented with green circles.</p>
Full article ">Figure 6
<p>PMF7G02448 (MYB family) directly binds to the promoter of three PmCesA genes. (<b>a</b>) Frequencies of all 10-mers are compared between the SELEX library and the same library shuffled. The top 10 most enriched 10-mers are shown. (<b>b</b>) The de novo discovered motifs of PMF7G02448. The consensus sequences of mo1-mo6: mo1(YYYACCTAMTTTN), mo2(NYYACCWAMCNN), mo2(NNRKTWGGTRTACCWAMYNN), mo2(NNNACCWAMYNNNNYACCWAMNH), mo2(NNNACCWAMYNYACCWAMNN), mo2(NNNACCWAMYNYYYACCWAMNH). (<b>c</b>) Enrichment of each motif in the SELEX library, evaluated by the fold change in motif matches between the SELEX and the shuffled libraries. (<b>d</b>) The affinity of genomic sequences to PMF7G02448 is visualized around the three <span class="html-italic">PmCesA</span> genes. Red arrows indicate the high affinity sites of PMF7G02448 in the promoter region (transcription start site (TSS) + 2kb). Affinity is defined by the SELEX enrichment scores of 10-mers.</p>
Full article ">
29 pages, 5564 KiB  
Review
Synthesis and Properties of α-Phosphate-Modified Nucleoside Triphosphates
by Alina I. Novgorodtseva, Alexander A. Lomzov and Svetlana V. Vasilyeva
Molecules 2024, 29(17), 4121; https://doi.org/10.3390/molecules29174121 - 30 Aug 2024
Viewed by 913
Abstract
This review article is focused on the progress made in the synthesis of 5′-α-P-modified nucleoside triphosphates (α-phosphate mimetics). A variety of α-P-modified nucleoside triphosphates (NTPαXYs, Y = O, S; X = S, Se, BH3, alkyl, amine, N-alkyl, imido, or others) have [...] Read more.
This review article is focused on the progress made in the synthesis of 5′-α-P-modified nucleoside triphosphates (α-phosphate mimetics). A variety of α-P-modified nucleoside triphosphates (NTPαXYs, Y = O, S; X = S, Se, BH3, alkyl, amine, N-alkyl, imido, or others) have been developed. There is a unique class of nucleoside triphosphate analogs with different properties. The main chemical approaches to the synthesis of NTPαXYs are analyzed and systematized here. Using the data presented here on the diversity of NTPαXYs and their synthesis protocols, it is possible to select an appropriate method for obtaining a desired α-phosphate mimetic. Triphosphates’ substrate properties toward nucleic acid metabolism enzymes are highlighted too. We reviewed some of the most prominent applications of NTPαXYs including the use of modified dNTPs in studies on mechanisms of action of polymerases or in systematic evolution of ligands by exponential enrichment (SELEX). The presence of heteroatoms such as sulfur, selenium, or boron in α-phosphate makes modified triphosphates nuclease resistant. The most distinctive feature of NTPαXYs is that they can be recognized by polymerases. As a result, S-, Se-, or BH3-modified phosphate residues can be incorporated into DNA or RNA. This property has made NTPαXYs a multifunctional tool in molecular biology. This review will be of interest to synthetic chemists, biochemists, biotechnologists, or biologists engaged in basic or applied research. Full article
(This article belongs to the Special Issue Chemistry of Nucleosides and Nucleotides and Their Analogues)
Show Figures

Scheme 1

Scheme 1
<p>The scheme of enzymatic synthesis of oligonucleotides from α-phosphate-modified nucleoside triphosphates.</p>
Full article ">Scheme 2
<p>Synthesis of thymidine 5′-(α-P-thio)triphosphate <b>1</b> [<a href="#B19-molecules-29-04121" class="html-bibr">19</a>].</p>
Full article ">Scheme 3
<p>Synthesis of adenosine 5′-(α-P-thio)triphosphate <b>2</b> [<a href="#B20-molecules-29-04121" class="html-bibr">20</a>].</p>
Full article ">Scheme 4
<p>Synthesis of adenosine 5′-(α-P-thio)triphosphate <b>2</b> [<a href="#B21-molecules-29-04121" class="html-bibr">21</a>].</p>
Full article ">Scheme 5
<p>Synthesis of 5′-(α-P-thio)triphosphate or 5′-(α-P-thio)(β,γ-methylene)triphosphate RNA <b>3</b> on a solid support. Piv = pivaloyl, CNE = cyanoethyl, DBU = 1,8-diazabicyclo[5.4.0]undec-7-ene, A<sup>Pac</sup> = N-6-(phenoxyacetyl)adenine, G<sup>iPrPac</sup> = N-2-(isopropylphenoxyacetyl)guanine, and C<sup>Ac</sup> = N-4-(acetyl)cytosine [<a href="#B24-molecules-29-04121" class="html-bibr">24</a>].</p>
Full article ">Scheme 6
<p>Synthesis of 5′-(α-P-thio)triphosphates of adenosine, cytidine, guanosine, and thymidine <b>6</b> [<a href="#B25-molecules-29-04121" class="html-bibr">25</a>].</p>
Full article ">Scheme 7
<p>Synthesis of nucleoside 5′-(α-P-thio)triphosphate <b>11</b> with a cyclic intermediate. R<sub>1</sub>: a protected hydroxyl group, and R<sub>2</sub>: H or OAc [<a href="#B27-molecules-29-04121" class="html-bibr">27</a>].</p>
Full article ">Scheme 8
<p>Synthesis of 5′-triphosphates with α-P-thio substitution <b>15</b> or α-P-selenium substitution <b>16</b> from unprotected nucleosides [<a href="#B28-molecules-29-04121" class="html-bibr">28</a>].</p>
Full article ">Scheme 9
<p>Synthesis of thymidine-(P-α-borano, P-α-thio)triphosphate <b>21</b> [<a href="#B30-molecules-29-04121" class="html-bibr">30</a>].</p>
Full article ">Scheme 10
<p>Synthesis of 3′-azidothymidine 5′-γ-P-propargylamido(α-P-S or α-P-Se) derivatives of triphosphate <b>26a</b>–<b>c</b> [<a href="#B31-molecules-29-04121" class="html-bibr">31</a>].</p>
Full article ">Scheme 11
<p>Synthesis of 5′-(α-P-S, α-P-Se, and α-P-BH<sub>3</sub>) derivatives of triphosphate <b>27a</b>–<b>c</b> by the amidophosphite method [<a href="#B10-molecules-29-04121" class="html-bibr">10</a>].</p>
Full article ">Scheme 12
<p>Synthesis of thymidine 5ʹ-(α-P-borano)triphosphate <b>30</b> by the amidophosphite method [<a href="#B32-molecules-29-04121" class="html-bibr">32</a>].</p>
Full article ">Scheme 13
<p>Synthesis of boranotriphosphate <b>32</b> by the amidophosphite method [<a href="#B33-molecules-29-04121" class="html-bibr">33</a>].</p>
Full article ">Scheme 14
<p>Synthesis of acyclothymidine 5′-(α-P-borano)triphosphate <b>33</b> [<a href="#B34-molecules-29-04121" class="html-bibr">34</a>].</p>
Full article ">Scheme 15
<p>Synthesis of 5′-(α-P-borano)triphosphates of ribonucleosides (A, U, G, and C) <b>35</b> by the one-pot procedure [<a href="#B35-molecules-29-04121" class="html-bibr">35</a>].</p>
Full article ">Scheme 16
<p>Synthesis of AZT 5′-triphosphate mimics <b>36a</b>–<b>c</b> [<a href="#B38-molecules-29-04121" class="html-bibr">38</a>].</p>
Full article ">Scheme 17
<p>The scheme of the synthesis of thymidine 5′-(α-P-methyl)triphosphate <b>37</b> [<a href="#B39-molecules-29-04121" class="html-bibr">39</a>].</p>
Full article ">Scheme 18
<p>The scheme of chemoenzymatic synthesis of 5′-(α-P-methyl)triphosphates of nucleosides <b>38a</b>–<b>c</b> [<a href="#B41-molecules-29-04121" class="html-bibr">41</a>].</p>
Full article ">Scheme 19
<p>Synthesis of 5′-(α-P-methyl)triphosphate of thymidine and of 3′-azidothymidine <b>41a</b>–<b>c</b> [<a href="#B42-molecules-29-04121" class="html-bibr">42</a>].</p>
Full article ">Scheme 20
<p>The scheme for the preparation of thymidine 5′-(α-P-amido)triphosphate and adenosine 5′-(α-P-amido)triphosphate [<a href="#B44-molecules-29-04121" class="html-bibr">44</a>].</p>
Full article ">Scheme 21
<p>Synthesis of 5′-(α-P-amido)triphosphates and 5′-(α-P-alkylamido)triphosphates of nucleosides [<a href="#B46-molecules-29-04121" class="html-bibr">46</a>].</p>
Full article ">Scheme 22
<p>The scheme of the preparation of 2′-deoxythymidine 5′-(α-P-(4-acetamidobenzenesulfonyl)imido)triphosphate via a cyclotriphosphite intermediate [<a href="#B48-molecules-29-04121" class="html-bibr">48</a>]. TEA: triethylamine.</p>
Full article ">Scheme 23
<p>The scheme of the synthesis of adenosine and thymidine 5′-(α-P-dimethylimidazolyl)monophosphates [<a href="#B49-molecules-29-04121" class="html-bibr">49</a>].</p>
Full article ">Scheme 24
<p>Resonance structures of 2′-deoxynucleotides carrying a dimethylimidazole substituent. A: Imido monophosphate; and B: amido monophosphate.</p>
Full article ">Scheme 25
<p>The scheme of obtaining thymidine 5′-(α-P-(1,3-dimethylimidazolidin-2-ylidene)triphosphate (TTPαDMI) <b>50</b> [<a href="#B49-molecules-29-04121" class="html-bibr">49</a>]. N-MeIm: N-methylimidazole.</p>
Full article ">Scheme 26
<p>Structures of diastereomers of phosphorothioate nucleosides and oligonucleotides.</p>
Full article ">
19 pages, 5361 KiB  
Review
Aptamer Screening: Current Methods and Future Trend towards Non-SELEX Approach
by Zhihui Fang, Xiaorui Feng, Fan Tang, Han Jiang, Shuyuan Han, Ran Tao and Chenze Lu
Biosensors 2024, 14(7), 350; https://doi.org/10.3390/bios14070350 - 18 Jul 2024
Cited by 1 | Viewed by 2068
Abstract
Aptamers are nucleic acid sequences that specifically bind with target molecules and are vital to applications such as biosensing, drug development, disease diagnostics, etc. The traditional selection procedure of aptamers is based on the Systematic Evolution of Ligands by an Exponential Enrichment (SELEX) [...] Read more.
Aptamers are nucleic acid sequences that specifically bind with target molecules and are vital to applications such as biosensing, drug development, disease diagnostics, etc. The traditional selection procedure of aptamers is based on the Systematic Evolution of Ligands by an Exponential Enrichment (SELEX) process, which relies on repeating cycles of screening and amplification. With the rapid development of aptamer applications, RNA and XNA aptamers draw more attention than before. But their selection is troublesome due to the necessary reverse transcription and transcription process (RNA) or low efficiency and accuracy of enzymes for amplification (XNA). In light of this, we review the recent advances in aptamer selection methods and give an outlook on future development in a non-SELEX approach, which simplifies the procedure and reduces the experimental costs. We first provide an overview of the traditional SELEX methods mostly designed for screening DNA aptamers to introduce the common tools and methods. Then a section on the current screening methods for RNA and XNA is prepared to demonstrate the efforts put into screening these aptamers and the current difficulties. We further predict that the future trend of aptamer selection lies in non-SELEX methods that do not require nucleic acid amplification. We divide non-SELEX methods into an immobilized format and non-immobilized format and discuss how high-resolution partitioning methods could facilitate the further improvement of selection efficiency and accuracy. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The schematic diagrams of SELEX methods in non-immobilized format. (<b>a</b>): The principle of CE-SELEX method. The method isolates substances based on electrophoretic mobility. Nucleic acid libraries are incubated with targets to form an equilibrium mixture. The components are separated due to difference in mobility in electric field [<a href="#B18-biosensors-14-00350" class="html-bibr">18</a>]; (<b>b</b>): The principle of cell-SELEX method. The target molecules on the cell surface are in their native state and do not require additional purification sequences that bind with these target molecules in their natural conformation [<a href="#B19-biosensors-14-00350" class="html-bibr">19</a>]; (<b>c</b>): the principle of GO-SELEX method, ssDNA already bound to targets will not be adsorbed by GO due to the change of spatial structure, which allows separation of potential aptamer sequences [<a href="#B20-biosensors-14-00350" class="html-bibr">20</a>].</p>
Full article ">Figure 2
<p>The schematic diagrams of SELEX methods in immobilized format. (<b>a</b>): The principle of capture-SELEX method. The complementary chains of the docking sequences in the library are immobilized on magnetic beads. After mixing and incubation, the conformation of the target binding sequences changes and can be released from the magnetic beads [<a href="#B21-biosensors-14-00350" class="html-bibr">21</a>]; (<b>b</b>): The principle of AFM-SELEX method. A nucleic acid library is immobilized on the gold chip and the target is functionalized on the microcantilever. The aptamers is released from the gold chip when the binding affinity is high enough [<a href="#B22-biosensors-14-00350" class="html-bibr">22</a>]; (<b>c</b>): The principle of affinity chromatography SELEX. The aptamers are immobilized on the surface of the magnetic beads and loaded into the affinity column. The targets interact specifically with the aptamers when samples are injected, and potential aptamers are selectively adsorbed onto the column and eluted for further analysis [<a href="#B23-biosensors-14-00350" class="html-bibr">23</a>]; (<b>d</b>): Microarray-SELEX divides multiple regions on the surface of a chip, each of which can point to a target, and then the library is added to the chip and screened for specific aptamers through multiple rounds of positive and negative selection [<a href="#B24-biosensors-14-00350" class="html-bibr">24</a>].</p>
Full article ">Figure 3
<p>The schematic diagrams of aptamer screening methods independent of intermediate nucleic acid amplification. (<b>a</b>): The principle of magnetic bead-assisted screening. The target is immobilized on magnetic beads and incubated with the addition of the nucleic acid library, then the bound and unbound aptamers are separated with a magnet. The magnetic beads are collected using a magnetic rack, and the sequences bound to the target are eluted and added to the next microcentrifuge tube containing fresh target-coupled magnetic beads for a new round of screening. The aptamers from the final round of screening are incubated with naked magnetic beads to exclude sequences that are specifically bound to the beads [<a href="#B64-biosensors-14-00350" class="html-bibr">64</a>]; (<b>b</b>): The principle of competition-enhanced ligand screening. The random nucleic acid library is divided into several aliquots and then one of the aliquots is taken first and added to the target. After incubation, the unbound sequences are washed and removed. A new aliquot of the library is then added to the target–DNA complex and the newly added sequence competes with the bound sequences for the target [<a href="#B65-biosensors-14-00350" class="html-bibr">65</a>]; (<b>c</b>): The principle of centrifugal distribution method. After incubation of the random nucleic acid library with the target, a high-speed centrifugal filter is used to separate the complex from the free oligonucleotide. Negative selection and positive selection are repeated alternately to obtain specific sequences [<a href="#B66-biosensors-14-00350" class="html-bibr">66</a>]; (<b>d</b>): The principle of fluorescence-activated cell sorting. Cells are specifically labeled with fluorescent dyes and then the fluorescent signals are excited by the laser of a flow cytometer to classify and sort the cells according to different fluorescent signal intensities. This technique allows for large-scale screening [<a href="#B49-biosensors-14-00350" class="html-bibr">49</a>].</p>
Full article ">
27 pages, 2298 KiB  
Review
New Insights into Aptamers: An Alternative to Antibodies in the Detection of Molecular Biomarkers
by Michaela Domsicova, Jana Korcekova, Alexandra Poturnayova and Albert Breier
Int. J. Mol. Sci. 2024, 25(13), 6833; https://doi.org/10.3390/ijms25136833 - 21 Jun 2024
Cited by 5 | Viewed by 3335
Abstract
Aptamers are short oligonucleotides with single-stranded regions or peptides that recently started to transform the field of diagnostics. Their unique ability to bind to specific target molecules with high affinity and specificity is at least comparable to many traditional biorecognition elements. Aptamers are [...] Read more.
Aptamers are short oligonucleotides with single-stranded regions or peptides that recently started to transform the field of diagnostics. Their unique ability to bind to specific target molecules with high affinity and specificity is at least comparable to many traditional biorecognition elements. Aptamers are synthetically produced, with a compact size that facilitates deeper tissue penetration and improved cellular targeting. Furthermore, they can be easily modified with various labels or functional groups, tailoring them for diverse applications. Even more uniquely, aptamers can be regenerated after use, making aptasensors a cost-effective and sustainable alternative compared to disposable biosensors. This review delves into the inherent properties of aptamers that make them advantageous in established diagnostic methods. Furthermore, we will examine some of the limitations of aptamers, such as the need to engage in bioinformatics procedures in order to understand the relationship between the structure of the aptamer and its binding abilities. The objective is to develop a targeted design for specific targets. We analyse the process of aptamer selection and design by exploring the current landscape of aptamer utilisation across various industries. Here, we illuminate the potential advantages and applications of aptamers in a range of diagnostic techniques, with a specific focus on quartz crystal microbalance (QCM) aptasensors and their integration into the well-established ELISA method. This review serves as a comprehensive resource, summarising the latest knowledge and applications of aptamers, particularly highlighting their potential to revolutionise diagnostic approaches. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>): Primary sequence of an aptamer is folded into a 3D structure to recognize its target. B to E represent options of 3D structural configurations: (<b>B</b>) a hairpin, (<b>C</b>) pseudoknot, (<b>D</b>) joined hairpins, and (<b>E</b>) quadruplex.</p>
Full article ">Figure 2
<p>A schematic illustration of the SELEX process for identifying novel target-specific aptamers. This process can be carried out in vitro.</p>
Full article ">Figure 3
<p>A schematic of the enzyme-linked aptamer-sorbent assay shows different approaches: direct ELASA, indirect ELASA, and sandwich ELASA. In direct ELASA, the aptamer can be directly conjugated to an enzyme or another reporter molecule. In indirect ELASA, the aptamer is first labelled with a reporter molecule such as biotin and then forms a complex with a secondary antibody–enzyme conjugate. The sandwich ELASA involves binding a biotinylated aptamer to capture the target antigen on a surface; subsequently, it uses the same aptamer-reporter molecule and secondary antibody pairing to create a colorimetric change.</p>
Full article ">Figure 4
<p>The principle of QCM aptasensor detection is illustrated in the graph, which shows the frequency changes in response to binding events. The graph shows the following stages: I—the initial frequency of the QCM sensor with an immobilised recognition element, which is an aptamer; II—the binding of target molecules; III—the binding of labelled secondary molecules; and IV—the regeneration of the sensor surface.</p>
Full article ">
16 pages, 1330 KiB  
Review
Therapeutic Applications of Aptamers
by George Santarpia and Eric Carnes
Int. J. Mol. Sci. 2024, 25(12), 6742; https://doi.org/10.3390/ijms25126742 - 19 Jun 2024
Cited by 1 | Viewed by 1552
Abstract
Affinity reagents, or target-binding molecules, are quite versatile and are major workhorses in molecular biology and medicine. Antibodies are the most famous and frequently used type and they have been used for a wide range of applications, including laboratory techniques, diagnostics, and therapeutics. [...] Read more.
Affinity reagents, or target-binding molecules, are quite versatile and are major workhorses in molecular biology and medicine. Antibodies are the most famous and frequently used type and they have been used for a wide range of applications, including laboratory techniques, diagnostics, and therapeutics. However, antibodies are not the only available affinity reagents and they do have significant drawbacks, including laborious and costly production. Aptamers are one potential alternative that have a variety of unique advantages. They are single stranded DNA or RNA molecules that can be selected for binding to many targets including proteins, carbohydrates, and small molecules—for which antibodies typically have low affinity. There are also a variety of cost-effective methods for producing and modifying nucleic acids in vitro without cells, whereas antibodies typically require cells or even whole animals. While there are also significant drawbacks to using aptamers in therapeutic applications, including low in vivo stability, aptamers have had success in clinical trials for treating a variety of diseases and two aptamer-based drugs have gained FDA approval. Aptamer development is still ongoing, which could lead to additional applications of aptamer therapeutics, including antitoxins, and combinatorial approaches with nanoparticles and other nucleic acid therapeutics that could improve efficacy. Full article
Show Figures

Figure 1

Figure 1
<p>Structures of aptamer–ligand complexes. (<b>A</b>) Aptamer in complex with theophylline (yellow) and magnesium (green) and sodium (purple) ions. Light blue dashed lines represent hydrogen bonding, darker dashed lines indicate ionic bonding. PDB: 8D28. Aptamers are also shown in complex with <span class="html-italic">Bacillus anthracis</span> ribosomal protein S8 (<b>B</b>; PDB: 4PDB) and TetR (<b>C</b>; PDB: 6SY6), both shown in blue.</p>
Full article ">Figure 2
<p>Graphical illustration of SELEX procedure with example methodologies and variations are included at each step. Red and blue oligos represent target-binding aptamers, while all other colors represent non-binding oligos.</p>
Full article ">
16 pages, 2617 KiB  
Article
Aptamer and Electrochemical Aptasensor towards Selenate Ions (SeO42−)
by Anna Szymczyk, Martyna Popiołek, Dominika Baran, Marcin Olszewski, Robert Ziółkowski and Elżbieta Malinowska
Int. J. Mol. Sci. 2024, 25(12), 6660; https://doi.org/10.3390/ijms25126660 - 17 Jun 2024
Viewed by 782
Abstract
Selenium is an essential inorganic compound in human and animal nutrition, involved in the proper functioning of the body. As a micronutrient, it actively contributes to the regulation of various metabolic activities, i.e., thyroid hormone, and protection against oxidative stress. However, Se exhibits [...] Read more.
Selenium is an essential inorganic compound in human and animal nutrition, involved in the proper functioning of the body. As a micronutrient, it actively contributes to the regulation of various metabolic activities, i.e., thyroid hormone, and protection against oxidative stress. However, Se exhibits a narrow concentration window between having a positive effect and exerting a toxic effect. In higher doses, it negatively affects living organisms and causes DNA damage through the formation of free radicals. Increased reactivity of Se anions can also disrupt the integrity and function of DNA-repairing proteins. As the permissible concentration of Se in drinking water is 10 µg/L, it is vital to develop sensitive and robust methods of Se detection in aqueous samples. In this study, for the first time, we proposed a selective aptamer for selenate ion detection, chosen following the SELEX process, and its application in the construction of an electrochemical aptasensor towards SeO42− ions. Measurement conditions such as the used redox marker and pH value of the measurement solution were chosen. The proposed aptasensor is characterized by good selectivity and an LOD of 1 nM. Conditions for biosensor regeneration and storage were also investigated in this research. Full article
(This article belongs to the Special Issue Recent Advances on Bioreceptors and Nanomaterial-Based Biosensors)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the biosensor response mechanism and the equation used for biosensor response calculation.</p>
Full article ">Figure 2
<p>SWV voltammograms. (<b>A</b>–<b>C</b>): Biosensor response registered with methylene blue as a redox marker with an increasing selenate ion concentration (from 1 to 12 μM); (<b>D</b>): biosensor response registered with ferro-/ferricyanide as a redox marker with a selenate ion concentration of 100 μM.</p>
Full article ">Figure 3
<p>CV voltammogram for biosensor response registered with methylene blue as a redox marker with a selenate ion concentration of 12 μM.</p>
Full article ">Figure 4
<p>Dependency between selenate ion concentration and calculated biosensor response (result obtained for SWV experiments, <span class="html-italic">n</span> = 4).</p>
Full article ">Figure 5
<p>SWV voltammograms for biosensor response registered with methylene blue (pH 4.0) as a redox marker and for selenate ion concentration at the level of (<b>A</b>) 1 nM and (<b>B</b>) 50 nM.</p>
Full article ">Figure 6
<p>(<b>A</b>) Dependency between selenate ion concentration and calculated biosensor response (result obtained for SWV experiments, <span class="html-italic">n</span> = 4); (<b>B</b>) plot of the absolute aptasensor electrochemical responses (transformed to positive values versus calculated with the equation presented in <a href="#ijms-25-06660-f001" class="html-fig">Figure 1</a>) in the function of analyte concentration for the determination of equilibrium dissociation constant (K<sub>D</sub>). Black points represent experimental points; red curve represents fitting through non-linear regression according to the “one-to-one” kinetic model.</p>
Full article ">Figure 7
<p>Selectivity studies of as-prepared electrochemical aptasensor. All ions were at the level of 100 nM and the sensor incubation in the sample was conducted for 30 min (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 8
<p>Response of the electrodes modified with appropriate electrode blocking agent (cysteamine, 6-mercapto-1-hexanol, 11-mercaptoundecanoic acid or 1-pentanethiol) for measurement solution containing only methylene blue (black line) and for methylene solution with selenate ion addition (100 nM) (red line).</p>
Full article ">Figure 9
<p>Real sample analysis with the use of the prepared biosensor. The selenate ion concentration was 100 nM (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 10
<p>(<b>A</b>) Aptasensor regeneration studies. Black line: before selenate ion detection; red line: after 30 min of electrode incubation in selenate ion solution (100 nM); blue line: after 20 min of aptasensor incubation in 0.5 M Na<sub>2</sub>CO<sub>3</sub> at pH 10.5 (regeneration); green line: after 30 min of electrode incubation in selenate ion solution (after regeneration process); (<b>B</b>) aptasensor response (50 nM selenate ion concentration) after one week of storage in the specific conditions (blue bar: response obtained with freshly prepared biosensor).</p>
Full article ">
15 pages, 3425 KiB  
Article
In Vivo Evaluation of 68Ga-Labeled NOTA-EGFRvIII Aptamer in EGFRvIII-Positive Glioblastoma Xenografted Model
by Jun Young Park, Ye Lim Cho, Tae Sup Lee, Daekyun Lee, Ju-Hyung Kang, Soryong Lim, Yujin Lee, Jae Hyun Lim and Won Jun Kang
Pharmaceutics 2024, 16(6), 814; https://doi.org/10.3390/pharmaceutics16060814 - 16 Jun 2024
Viewed by 1214
Abstract
EGFRvIII is expressed only in tumor cells and strongly in glioblastoma and is considered a promising target in cancer diagnosis and therapy. Aptamers are synthetic single-stranded oligonucleotides that bind to biochemical target molecules with high binding affinity and specificity. This study examined the [...] Read more.
EGFRvIII is expressed only in tumor cells and strongly in glioblastoma and is considered a promising target in cancer diagnosis and therapy. Aptamers are synthetic single-stranded oligonucleotides that bind to biochemical target molecules with high binding affinity and specificity. This study examined the potential of the 68Ga-NOTA-EGFRvIII aptamer as a nuclear imaging probe for visualizing EGFRvIII-expressing glioblastoma by positron emission tomography (PET). EGFRvIII aptamer was selected using the SELEX technology, and flow cytometry and fluorescence microscopy verified the high binding affinity to EGFRvIII positive U87MG vIII 4.12 glioma cells but not to EGFRvIII negative U87MG cells. The EGFRvIII aptamer was conjugated with a chelator (1,4,7-triazanonane-1,4,7-triyl)triacetic acid (NOTA) for 68Ga-labeling. The 68Ga-NOTA-EGFRvIII aptamer was prepared using the preconcentration-based labeling method with a high radiolabeling yield at room temperature. Ex vivo biodistribution analyses confirmed the significantly higher tumor uptake of the 68Ga-NOTA-EGFRvIII aptamer in EGFRvIII-expressing xenograft tumors than that in EGFRvIII negative tumors, confirming the specific tumor uptake of the 68Ga-NOTA-EGFRvIII aptamer in vivo. PET imaging studies revealed a high retention rate of the 68Ga-NOTA-EGFRvIII aptamer in U87MG vIII 4.12 tumors but only low uptake levels in U87-MG tumors, suggesting that the 68Ga-NOTA-EGFRvIII aptamer may be used as a PET imaging agent for EGFRvIII-expressing glioblastoma. Full article
(This article belongs to the Special Issue Advances in Radiopharmaceuticals for Disease Diagnoses and Therapy)
Show Figures

Figure 1

Figure 1
<p>Evaluation of EGFRvIII expression in glioma cell lines. (<b>A</b>) EGFRvIII expression in the EGFRvIII-engineered U87MG cell lines (DKMG/EGFRvIII and U87MG vIII 4.12) and U87MG cells was analyzed by flow cytometry using a monoclonal antibody (mAb) against EGFRvIII. (<b>B</b>) The Western blot analysis of the EGFRvIII level in the U87MG vIII 4.12 and U87MG cells. β-Actin was used as an endogenous control. (<b>C</b>) The relative intensities of the EGFRvIII proteins were calculated by comparing them to the intensity of β-actin. The data are presented as the mean ± SD of three independent experiments. Statistical analysis was performed using a Student’s <span class="html-italic">t</span>-test. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Characterization of the EGFRvIII aptamer in vitro. (<b>A</b>) The secondary structure of the EGFRvIII aptamer predicted by the RNAstructure web server according to the free energy minimization algorithm. (<b>B</b>) The flow cytometry analysis of the binding ability of the Cy5-labeled EGFRvIII aptamer (Cy5-EGFRvIII) against the EGFRvIII-positive U87MG vIII 4.12 cells and EGFRvIII-negative U87MG cells at 4 °C. The Cy5-labeled scrambled EGFRvIII (Cy5-ScrEGFRvIII) aptamer was used as the negative control. (<b>C</b>) The fluorescent microscopy of cell binding by the Cy5-EGFRvIII aptamer (red) to the U87MG vIII 4.12 cells and U87MG cells. The cells were stained with the Cy5-EGFRvIII aptamer at 4 °C, followed by washing and confocal microscopy imaging. The nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI, blue). Scale bar = 100 µm.</p>
Full article ">Figure 3
<p>Characterization of the NOTA-EGFRvIII aptamer. (<b>A</b>) The schematic diagram of synthesis process for the preparation of the (1,4,7-triazanonane-1,4,7-triyl)triacetic acid (NOTA)-conjugated EGFRvIII (NOTA-EGFRvIII) aptamer. (<b>B</b>) The biolayer interferometry (BLI) data of the EGFRvIII aptamers and NOTA-EGFRvIII aptamers against various concentrations of EGFRvIII protein. (<b>C</b>) The evaluation of EGFRvIII aptamer conjugation with NOTA using urea/polyacrylamide gel electrophoresis. Lane M: 20/100 DNA ladder, Lane 1: empty vector, Lane 2: EGFRvIII aptamer, and Lane 3: NOTA-EGFRvIII aptamer.</p>
Full article ">Figure 4
<p>Radiolabeling of the NOTA-EGFRvIII aptamer with gallium-68. (<b>A</b>) The schematic diagram of the anionic purification of the <sup>68</sup>Ga eluate and the synthesis of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer. (<b>B</b>) The representative radio-TLC chromatograms of the <sup>68</sup>Ga-NOTA-EGFRvIII (<b>upper</b> image) and free <sup>68</sup>Ga<sup>3+</sup> (<b>lower</b> image). R<sub>f</sub>: relative to the front. (<b>C</b>) The influence of the reaction time on the <sup>68</sup>Ga-radiolabeling of the NOTA-EGFRvIII aptamer. The radiolabeling was analyzed by radio-TLC. (<b>D</b>) The in vitro stability of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer was investigated in PBS buffers at 37 °C. The radiochemical purity of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer was monitored by radio-TLC for up to 60 min. The data represent the mean ± SD from three independent experiments.</p>
Full article ">Figure 5
<p>Ex vivo biodistribution of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer in the mice bearing subcutaneous U87MG vIII 4.12 tumors. The uptake levels of the blood, heart, liver, lung, spleen, stomach, muscle, small intestine (S.I.), large intestine (L.I.), bone, and tumor at 30 min and 60 min after the tail vein injection of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer in the mice bearing subcutaneous (<b>A</b>) EGFRvIII-positive U87MG vIII 4.12 tumors or (<b>B</b>) EGFRvIII-negative U87MG tumors. The whole biodistribution profiles are shown in the <a href="#app1-pharmaceutics-16-00814" class="html-app">Supplementary Material</a>. The data are expressed as a percentage of injected dose per gram tissue (%ID/g ± SD, n = 4 at each time point).</p>
Full article ">Figure 6
<p>MicroPET imaging of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer in the mice bearing subcutaneous U87MG vIII 4.12 tumors. (<b>A</b>) Representative coronal and transverse microPET images at 60 min after a tail vein injection of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer (<b>left</b>) or <sup>68</sup>Ga<sup>3+</sup> (<b>right</b>). The white arrows indicate the location of the tumors. (<b>B</b>) The representative whole-body microPET images of the <sup>68</sup>Ga-NOTA-EGFRvIII aptamer in the mice bearing the EGFRvIII-positive U87MG vIII 4.12 (<b>right</b>) and EGFRvIII negative U87MG tumors (<b>left</b>). The white arrow indicated the location of the U87MG vIII 4.12 tumors, and the red arrow indicated the region of the U87MG tumors.</p>
Full article ">
12 pages, 5696 KiB  
Article
Development of Aptamers for RNase Inactivation in Xtract-Free™ Sample Collection and Transport Medium
by Luke T. Daum, John D. Rodriguez and James P. Chambers
Diagnostics 2024, 14(12), 1207; https://doi.org/10.3390/diagnostics14121207 - 7 Jun 2024
Viewed by 1090
Abstract
There is a significant need to develop new environmentally friendly, extraction-free sample collection mediums that can effectively preserve and protect genetic material for point-of-care and/or self-collection, home-collection, and mail-back testing. Systematic evolution of ligands by exponential enrichment (SELEX) was used to create anti-ribonuclease [...] Read more.
There is a significant need to develop new environmentally friendly, extraction-free sample collection mediums that can effectively preserve and protect genetic material for point-of-care and/or self-collection, home-collection, and mail-back testing. Systematic evolution of ligands by exponential enrichment (SELEX) was used to create anti-ribonuclease (RNase) deoxyribonucleic acid (DNA) aptamers against purified RNase A conjugated to paramagnetic carboxylated beads. Following eight rounds of SELEX carried out under various stringency conditions, e.g., selection using Xtract-Free™ (XF) specimen collection medium and elevated ambient temperature of 28 °C, a panel of five aptamers was chosen following bioinformatic analysis using next-generation sequencing. The efficacy of aptamer inactivation of RNase was assessed by monitoring ribonucleic acid (RNA) integrity via fluorometric and real-time RT-PCR analysis. Inclusion of aptamers in reaction incubations resulted in an 8800- to 11,200-fold reduction in RNase activity, i.e., digestion of viral RNA compared to control. Thus, anti-RNase aptamers integrated into XF collection medium as well as other commercial reagents and kits have great potential for ensuring quality intact RNA for subsequent genomic analyses. Full article
(This article belongs to the Special Issue Microbiology Laboratory: Sample Collection and Diagnosis Advances)
Show Figures

Figure 1

Figure 1
<p>Overview of anti-RNase A aptamer production. Aptamer enrichment was performed using purified RNase A conjugated to paramagnetic carboxy beads that were incubated in Xtract-Free™ collection medium (LuJo Bioscience Laboratory, San Antonio, TX, USA) at an elevated (28 °C) temperature. The colors represent the approximately 10<sup>15</sup> different species of aptamers in the original SELEX pool.</p>
Full article ">Figure 2
<p>qRT-PCR results of enzymatic digestions using bead-RNase A conjugate. Pos CNTRL = reaction contains viral RNA and no RNase A (positive control); Carboxy beads = reaction contains viral RNA + carboxy beads (unbound); Bead-RNase conjugate = reaction contains viral RNA + bead-RNase conjugate. RNase A treated (no bead) = reaction contains viral RNA + equivalent RNase A (negative control). All reactions were incubated for 15 min @ 37 °C. Average real-time cycle qRT-PCR cycle threshold values from triplicate digestion reactions with standard deviation bars are shown.</p>
Full article ">Figure 3
<p>RNase A enzymatic activity using the RNaseAlert™ Substrate Nuclease Detection System (IDT, Coralville, IA, USA) from: (<b>A</b>) reactions assessed by Qubit fluorometer (Thermo Fisher, Waltham, MA, USA) measured every 30 s for 5 min and at 15 min, and (<b>B</b>) using ultraviolet light with visual inspection at 5 days. In (<b>A</b>), reactions containing each aptamer (XF8 1-4 and XF8-F) were similar to Neg CNTRL reaction. Note: In (<b>B</b>), aptamer XF8-F is not shown but was similar to aptamers XF8-1 to 4, i.e., little to no visible fluorescence compared to control.</p>
Full article ">Figure 4
<p>Anti-RNase aptamer protection after bead-RNase conjugate incubation as assessed by qRT-PCR. Reactions containing aptamer were similar to positive control reaction containing no bead-RNase conjugate. Random DNA hexamers, a ssDNA non-aptamer (146 bp), and poly-A RNA were readily digested and like negative control reactions containing only viral RNA plus RNase A. All reactions were incubated for 15 min @ 37 °C. Average real-time qRT-PCR cycle threshold values from triplicate reactions with standard deviation bars are shown.</p>
Full article ">
Back to TopTop