Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mikrochim Acta ; 187(4): 251, 2020 03 30.
Article in English | MEDLINE | ID: mdl-32232575

ABSTRACT

A novel surface plasmon resonance (SPR) strategy is introduced for the specific determination of exosomes based on aptamer recognition and polydopamine-functionalized gold nanoparticle (Au@PDA NP)-assisted signal amplification. Exosomes derived from hepatic carcinoma SMMC-7721 were selected as the model target. SMMC-7721 exosomes can be specifically captured by the aptamer ZY-sls that was complementary to the DNA tetrahedron probes (DTPs), and then the CD63 aptamer-linked Au@PDA NPs recognized SMMC-7721 exosomes for signal amplification. The DTPs were modified on a Au film for preventing Au deposition on the surface during the introduction of HAuCl4, and PDA coated on the AuNPs was used to reduce HAuCl4 in situ without any reductant assistance. It results in a further enhanced SPR signal. The assay can clearly distinguish SMMC-7721 exosomes from others (HepG2 exosomes, Bel-7404 exosomes, L02 exosomes, MCF-7 exosomes, and SW480 exosomes, respectively). SMMC-7721 exosomes are specifically determined as low as 5.6 × 105 particles mL-1. The method has successfully achieved specific determination of SMMC-7721 exosomes even in 50% of human serum without any pretreatment. Graphical abstract A novel surface plasmon resonance (SPR) strategy was introduced for the specific determination of exosomes based on aptamer recognition and polydopamine functionalized gold nanoparticles (Au@PDA NPs). The SPR signal was improved using the Au@PDA NPs assisted amplification.


Subject(s)
Aptamers, Nucleotide/chemistry , Exosomes/chemistry , Indoles/chemistry , Metal Nanoparticles/chemistry , Polymers/chemistry , Cell Line, Tumor , Gold/chemistry , Humans , Surface Plasmon Resonance
2.
Anal Chem ; 92(3): 2853-2858, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31916749

ABSTRACT

The exhaustive investigating interactions between recognition probes and amyloid aggregates, especially simultaneous recognition events, are challenging and crucial for the design of biosensing probes and further diagnosis of amyloid diseases. In the present work, the interactions of aptamers (Apts) with ß-amyloid (Aß) aggregates were explored thoroughly by single-molecule force spectroscopy (SMFS). Indeed, it was found that the interaction of aptamer1 (Apt1)-amyloid aggregates was different from that of aptamer2 (Apt2)-Aß40 aggregates at the single-molecule level. Especially, the interaction force of Apt1-Aß40 fibril showed a double distinguishing Gaussian fitting. The only unimodal distribution of the force histogram was displayed for the interactions of Apt2-Aß40 oligomer, Apt2-Aß40 fibril, and Apt1-Aß40 oligomer. More intriguingly, two Apts could bind to amyloid aggregates simultaneously. With the assistance of two Apts recognition, a novel sensitive dual Apt-based surface plasmon resonance (SPR) sensor using Au nanoparticles (AuNPs) was developed for quantifying Aß40 aggregates. The dual Apt-based SPR sensor not only avoided the limitation of steric hindrance and epitope but also employed simple operation as well as inexpensive recognition probes. A detection limit as low as 0.2 pM for Aß40 oligomer and 0.05 pM for Aß40 fibril could be achieved. Moreover, the established sensor could be successfully applied to detect Aß40 aggregates in artificial cerebrospinal fluid (CSF) and undiluted real CSF. This work could provide a strategy to monitor a simultaneous recognition event using SMFS and broaden the application of Apts in the diagnosis of neurodegenerative diseases.


Subject(s)
Amyloid beta-Peptides/chemistry , Aptamers, Nucleotide/chemistry , Gold/chemistry , Microscopy, Atomic Force , Protein Aggregates
3.
ACS Appl Bio Mater ; 3(12): 8611-8618, 2020 Dec 21.
Article in English | MEDLINE | ID: mdl-35019631

ABSTRACT

Developing a strategy of modulating ß-amyloid (Aß) aggregation with low cost, easy synthesis, high efficiency, and biosafety is significant and a challenge for Alzheimer's disease (AD) therapy. Herein, DNA aptamer (Aß-Apt) against Aß42 obtained by in vitro selection was developed as a potent inhibitor of Aß42 aggregation for the first time. Indeed, the Aß42 monomer fibrillation was inhibited completely by Aß-Apt. Notably, the inhibition effect of Aß-Apt on the Aß42 oligomer aggregation was more obvious than that on the Aß42 monomer aggregation. It was presumed that the distinguishing effect may be attributed to different binding behaviors of Aß-Apt with Aß42 monomer and Aß42 oligomer. Surface plasmon resonance analysis demonstrated that Aß-Apt specifically recognized Aß42 monomer and Aß42 oligomer. Furthermore, the binding affinity of Aß-Apt with Aß42 oligomer was larger than that of Aß-Apt with Aß42 monomer. This work provided a promising platform with high efficiency for manipulating Aß aggregation.

4.
J Mol Recognit ; 33(5): e2829, 2020 05.
Article in English | MEDLINE | ID: mdl-31816660

ABSTRACT

Understanding the binding of split aptamer/its target could become a breakthrough in the application of split aptamer. Herein, vascular endothelial growth factor (VEGF), a major biomarker of human diseases, was used as a model, and its interaction with split aptamer was explored with single molecule force spectroscopy (SMFS). SMFS demonstrated that the interaction force of split aptamer/VEGF165 was 169.44 ± 6.59 pN at the loading rate of 35.2 nN/s, and the binding probability of split aptamer/VEGF165 was dependent on the concentration of VEGF165 . On the basis of dynamic force spectroscopy results, one activation barrier in the dissociation process of split aptamer/VEGF165 complexes was revealed, which was similar to that of the intact aptamer/VEGF165 . Besides, the dissociation rate constant (koff ) of split aptamer/VEGF165 was close to that of intact aptamer/VEGF165 , and the interaction force of split aptamer/VEGF165 was higher than the force of intact aptamer/VEGF165 . It indicated that split aptamer also possessed high affinity with VEGF165 . The work can provide a new method for exploring the interaction of split aptamer/its targets at single-molecule level.


Subject(s)
Microscopy, Atomic Force/methods , Vascular Endothelial Growth Factor A/metabolism , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Humans , Single Molecule Imaging
5.
ACS Appl Mater Interfaces ; 11(43): 40817-40825, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31556587

ABSTRACT

The real application of DNA-functionalized gold nanoparticles (DNA-Au NPs) was limited by decreased stability and irreversible aggregation in high-ionic strength solutions and complex systems. Therefore, exploring a kind of DNA-Au NPs with excellent stability in high-ionic strength solutions and complex systems is challenging and significant. Herein, a novel universal bioconjugate strategy for constructing ultrastable DNA-Au NPs was designed based on the combination of polydopamine (PDA) shell and DNA linker. The obtained DNA-linked Au@polydopamine nanoparticles (DNA-Au@PDA NPs) showed colloidal stability in high-ionic strength solution and complex systems (such as human serum and cell culture supernatant). Moreover, the nanoparticles still maintained good dispersion after multiple freeze-thaw cycles. The high stability of DNA-Au@PDA NPs may be attributed to increasing the electrostatic and steric repulsions among nanoparticles through the effect of both PDA shell and DNA linker on Au@PDA NPs. For investigating the application of such nanoparticles, a highly sensitive assay for miRNA 141 detection was developed using DNA-Au@PDA NPs coupled with dynamic light scattering (DLS). Comparing with the regular DNA-Au NPs, DNA-Au@PDA NPs could detect as low as 50 pM miRNA 141 even in human whole serum. Taken together, the features of Bio/Nanointerface make the nanoparticle suitable for various applications in harsh biological and environmental conditions due to the stability. This work may provide a universal modification method for obtaining stable nanoparticles.


Subject(s)
DNA/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , MicroRNAs/analysis , Humans , Indoles/chemistry , Materials Testing , Polymers/chemistry
6.
Anal Chem ; 91(12): 7943-7949, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31117415

ABSTRACT

Alkaline phosphatase (ALP) is a significant biomarker in clinical diagnostics, and the abnormal level of ALP enzyme in serum is closely related to various diseases such as bone or liver cancer, bone metastases, and extrahepatic biliary obstruction. Herein a simple and portable photothermal biosensor was developed for sensitive detection of ALP enzyme based on the formation of polydopamine (PDA) nanoparticles using a thermometer or temperature discoloration sticker as readout. A MnO2 nanosheet was first prepared using a novel one-pot strategy which was operationally simple and not overly time-consuming. Then dopamine (DA) was quickly polymerized into PDA nanoparticles in the presence of the MnO2 nanosheet. When the model analyte ALP was present, the substrate 2-phospho-l-ascorbic acid trisodium salt (AAP) was catalytically hydrolyzed into l-ascorbic acid (AA), resulting in the inhibition of the formation of the PDA nanoparticles owing to the fact that the MnO2 nanosheet was reduced to Mn2+ by the generated AA. Thus, a portable biosensor based on the photothermal properties of PDA nanoparticles for ALP enzyme detection was established with a detection limit as low as 0.1 U/L (thermometer) and 1 U/L (temperature discoloration sticker). In addition, it also showed excellent sensing performance for the ALP assay in human serum. Such a simple, label-free, cost-effective, and sensitive assay could exhibit real potential application for ALP detection and early diagnosis, especially in developing countries or remote regions.


Subject(s)
Alkaline Phosphatase/metabolism , Enzyme Assays/instrumentation , Point-of-Care Systems , Temperature , Thermometers , Animals , Cattle , Color , Indoles/chemistry , Manganese Compounds/chemistry , Nanoparticles/chemistry , Oxides/chemistry , Polymers/chemistry
7.
J Mol Recognit ; 31(4)2018 04.
Article in English | MEDLINE | ID: mdl-29143447

ABSTRACT

Increasing knowledge on the understanding interactions of aptamer with misfolded proteins (including monomer, oligomer, and amyloid fibril) is crucial for development of aggregation inhibitors and diagnosis of amyloid diseases. Herein, the interactions of lysozyme monomer-, oligomer-, and amyloid fibril-aptamer were investigated using single-molecule force spectroscopy. The results revealed that the aptamer screened against lysozyme monomer could also bind to oligomer and amyloid fibril, in spite of the recognition at a lower binding probability. It may be attributed to the inherent structural differences of misfolded proteins and the flexible conformation of aptamer. In addition, dynamic force spectra showed that there were similar dissociation paths in the dissociation process of lysozyme monomer-, oligomer-, and amyloid fibril-aptamer complexes. It showed that the dissociation only passed 1 energy barrier from the binding state to the detachment. However, the dynamic parameters suggested that the oligomer- and amyloid fibril-aptamer were more stable than lysozyme monomer-aptamer. The phenomena may result from the exposure of aptamer-recognized sequences on the surface and the electrostatic interactions. This work demonstrated that single-molecule force spectroscopy could be a powerful tool to study the binding behavior of the aptamer with misfolded proteins at single-molecule level, providing abundant information for researches and comprehensive applications of aptamer probes in diagnosis of amyloid diseases.


Subject(s)
Amyloid/metabolism , Aptamers, Nucleotide/metabolism , Single Molecule Imaging/methods , Amyloid/chemistry , Aptamers, Nucleotide/chemistry , Humans , Microscopy, Atomic Force , Models, Molecular , Molecular Conformation , Molecular Dynamics Simulation , Muramidase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...