Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(22): e111, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-37941139

RESUMEN

RNA In situ imaging through DNA self-assembly is advantaged in illustrating its structures and functions with high-resolution, while the limited reaction efficiency and time-consuming operation hinder its clinical application. Here, we first proposed a new strand displacement reaction (SDR) model (Cas12a thrusting SDR, CtSDR), in which Cas12a could overcome the inherent reaction limitation and dramatically enhance efficiency through energy replenishment and by-product consumption. The target-initiated CtSDR amplification was established for RNA analysis, with order of magnitude lower limit of detection (LOD) than the Cas13a system. The CtSDR-based RNA in situ imaging strategy was developed to monitor intra-cellular microRNA expression change and delineate the landscape of oncogenic RNA in 66 clinic tissue samples, possessing a clear advantage over classic in situ hybridization (ISH) in terms of operation time (1 h versus 14 h) while showing comparable sensitivity and specificity. This work presents a promising approach to developing advanced molecular diagnostic tools.


Asunto(s)
Técnicas Biosensibles , ARN , ARN/genética , Sistemas CRISPR-Cas , ADN/genética , ADN/química , Sensibilidad y Especificidad , Hibridación in Situ , Técnicas de Amplificación de Ácido Nucleico/métodos , Técnicas Biosensibles/métodos
2.
Anal Chem ; 96(5): 2253-2263, 2024 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-38277203

RESUMEN

Current study in the heterogeneity and physiological behavior of tumor cells is limited by the fluorescence in situ hybridization technology in terms of probe assembly efficiency, background suppression capability, and target compatibility. In a typically well-designed assay, hybridization probes are constructed in a confined nanostructure to achieve a rapid assembly for efficient signal response, while the excessively high local concentration between different probes inevitably leads to nonspecific background leakage. Inspired by the fabric zipper, we propose a novel confinement reaction pattern in a zipper-confined DNA nanoframe (ZCDN), where two kinds of hairpin probes are independently anchored respective tracks. The metastable states of the dual tracks can well avoid signal leakage caused by the nonspecific probe configuration change. Biomarker-mediated proximity ligation reduces the local distance of dual tracks, kinetically triggering an efficient allosteric chain reaction between the hairpin probes. This method circumvents nonspecific background leakage while maintaining a high efficiency in responding to targets. ZCDN is employed to track different cancer biomarkers located in both the cytoplasm and cytomembrane, of which the expression level and oligomerization behavior can provide crucial information regarding intratumoral heterogeneity. ZCDN exhibits high target response efficiency and strong background suppression capabilities and is compatible with various types of biological targets, thus providing a desirable tool for advanced molecular diagnostics.


Asunto(s)
Técnicas Biosensibles , Nanoestructuras , Hibridación Fluorescente in Situ , ADN/química , Diagnóstico por Imagen , Nanoestructuras/química , Sondas de ADN/genética , Sondas de ADN/química , Técnicas Biosensibles/métodos
3.
Anal Bioanal Chem ; 416(6): 1469-1483, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38236393

RESUMEN

This paper describes the simple and label-free detection of thrombin using optical fiber surface plasmon resonance (SPR) sensors based on gold films prepared by the cost-effective method of electroless plating. The plating conditions for simultaneously obtaining gold film on cylindrical core and end surfaces of an optical fiber suitable for measurement were optimized. The fabricated sensor exhibited a linear refractive index sensitivity of 2150 nm/RIU and 7.136 (a.u.)/RIU in the refractive index of 1.3329-1.3605 interrogated by resonance wavelength and amplitude methods respectively and a single wavelength monitoring method was proposed to investigate the sensing performance of this sensor. Polyadenine diblock and thiolated thrombin aptamers were immobilized on gold nanoparticles and gold films respectively to implement a sandwich optical fiber assay for thrombin. The developed optical fiber SPR sensors were successfully used in the determination of thrombin down to 0.56 nM over a wide range from 2 to 100 nM and showed good selectivity for thrombin, which indicated their potential clinical applications for biomedical samples.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , Resonancia por Plasmón de Superficie/métodos , Fibras Ópticas , Técnicas Biosensibles/métodos , Oro , Trombina
4.
Mikrochim Acta ; 191(3): 126, 2024 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-38332145

RESUMEN

An ultrasensitive sandwich-type electrochemical immunosensor for pro-gastrin-releasing peptide (ProGRP) detection was constructed based on PtCu nanodendrites functionalized Au/polyaniline nanospheres (Au/PANI@PtCu). The prepared Au/PANI@PtCu nanocomposites not only possessed excellent electro-catalytic activity of H2O2 reduction due to the synergistic effect between the Au/PANI and PtCu NDs but also provided large specific surface area for detection of antibodies (Ab2) immobilization. In addition, Au nanoparticles encapsulated multi-wall carbon nanotubes (AuNPs@MWCNTs) were also applied to modify the glassy carbon electrode interface for loading numerous capture antibodies (Ab1). In the presence of target ProGRP, a sandwich-type electrochemical immunosensor showed a strong current response from the electro-catalysis of Au/PANI@PtCu toward H2O2 reduction. Benefiting from the exceptional electro-catalytic performance of Au/PANI@PtCu and the high conductivity of AuNPs@MWCNTs, the sandwich-type immunoassay exhibited remarkable sensitivity in detection. The linear range extended from 100 fg/mL to 10 ng/mL, while achieving an impressively low limit of detection of 77.62 fg/mL.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , Nanotubos de Carbono , Péptido Liberador de Gastrina , Oro , Peróxido de Hidrógeno , Anticuerpos Inmovilizados , Inmunoensayo , Anticuerpos
5.
Anal Chem ; 95(9): 4486-4495, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36802524

RESUMEN

Exosomal microRNAs (exomiRNAs) have emerged as ideal biomarkers for early clinical diagnostics. The accurate detection of exomiRNAs plays a crucial role in facilitating clinical applications. Herein, an ultrasensitive electrochemiluminescent (ECL) biosensor was constructed using three-dimensional (3D) walking nanomotor-mediated CRISPR/Cas12a and tetrahedral DNA nanostructures (TDNs)-modified nanoemitters (TCPP-Fe@HMUiO@Au-ABEI) for exomiR-155 detection. Initially, the 3D walking nanomotor-mediated CRISPR/Cas12a strategy could effectively convert the target exomiR-155 into amplified biological signals for improving the sensitivity and specificity. Then, TCPP-Fe@HMUiO@Au nanozymes with excellent catalytic performance were used to amplify ECL signals because of the enhanced mass transfer and increased catalytic active sites, originating from its high surface areas (601.83 m2/g), average pore size (3.46 nm), and large pore volumes (0.52 cm3/g). Meanwhile, the TDNs as the scaffold to fabricate "bottom-up" anchor bioprobes could improve the trans-cleavage efficiency of Cas12a. Consequently, this biosensor achieved the limit of detection down to 273.20 aM ranging from 1.0 fM to 1.0 nM. Furthermore, the biosensor could discriminate breast cancer patients evidently by analyzing exomiR-155, and these results conformed to that of qRT-PCR. Thus, this work provides a promising tool for early clinical diagnostics.


Asunto(s)
Técnicas Biosensibles , MicroARNs , Humanos , MicroARNs/análisis , Sistemas CRISPR-Cas , ADN/química , Fotometría , Técnicas Biosensibles/métodos
6.
Anal Chem ; 95(5): 3098-3107, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36693787

RESUMEN

Nowadays, infections caused by methicillin-resistant Staphylococcus aureus (MRSA) have constituted a new challenge for anti-infective treatment. Precise identification and rapid clinical diagnostics of MRSA from other methicillin-sensitive strains entail assays with robust diagnostic efficiency and simple operation steps. Sensitive detection of MecA gene is promising to indicate MRSA infection, but it is challenged by the lack of isothermal and simple strategies. A visual assay based on isothermal rolling circular amplification and G-quadruplex/hemin (G4/hemin) DNAzyme proximity assembly was proposed for the immediate, efficient, and cost-effective detection of MecA in simple operation steps and in a single tube. The presence of MecA specifically drove the formation of circular templates, which further triggered isothermal amplification. The amplified product offered abundant binding sites for DNA-grafted hemin probes to form a novel proximity-assembled G4/hemin DNAzyme structure for colorimetric changing diagnosis. This tandem-repeated novel DNAzyme possessed higher catalytic activity and a lower background signal than traditional G4/hemin DNAzyme, ensuring sensitive discrimination of MRSA (limit of detection: 9.6 pM). Assay stability and antimatrix interference capability enable clinical application, which shows compared diagnostic ability with classic methods (100% sensitivity and 100% specificity) but possesses more simplified procedures and shorter turnaround time (<6 h). This colorimetric strategy in a nonsite-specific and hypersensitive manner holds foreseeable prospects in clinical diagnostic and research applications.


Asunto(s)
Técnicas Biosensibles , ADN Catalítico , G-Cuádruplex , Staphylococcus aureus Resistente a Meticilina , ADN Catalítico/química , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/metabolismo , Hemina/química , ADN , Técnicas Biosensibles/métodos
7.
Small ; 19(43): e2303034, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37365695

RESUMEN

Rapid and accurate imaging of the BCR/ABL fusion gene isoforms (e.g., e13a2, e14a2 and co-expression type) of chronic myeloid leukemia (CML) is of vital importance to first-line drug selection, but there is no assay that meets clinical needs (e.g., clinical kits > 18 h without isoforms information). Herein, an in situ imaging platform is developed for the rapid and accurate detection of CML fusion gene isoforms using asymmetric sequence-enhanced hairpins DNA encapsulated silver nanoclusters (ADHA) and catalyzed hairpin assembly (CHA). The specific detection of e13a2 and e14a2 fusion gene isoforms with detection limits of 19.2 am (11.558 copies µL-1 ) and 32.56 am (19.601 copies µL-1 ) in one-pot is achieved. The feasibility of the developed assay for real-world applications are demonstrated by one-step fluorescence imaging (40 min) of e13a2, e14a2 and co-expression type in bone marrow quantitatively (International Standard: 15.66%-168.878%) and further validated by cDNA-sequencing. This work suggests that the developed imaging platform holds great potential for rapid identification of the fusion gene isoforms and isoform related treatment monitoring.


Asunto(s)
Proteínas de Fusión bcr-abl , Leucemia Mielógena Crónica BCR-ABL Positiva , Humanos , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/uso terapéutico , Médula Ósea , Plata/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/diagnóstico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Isoformas de Proteínas/genética , ADN Complementario , Imagen Óptica
8.
Small ; 19(32): e2208142, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37066711

RESUMEN

Sensitive and specific analysis of extracellular vesicles (EVs) offers a promising minimally invasive way to identify malignant pulmonary nodules from benign lesions. However, accurate analysis of EVs is subject to free target proteins in blood samples, which compromises the clinical diagnosis value of EVs. Here a DNA-guided extracellular-vesicle metallization (DEVM) strategy is described for ultrasensitive and specific analysis of EV protein biomarkers and classification of pulmonary nodules. The facile DEVM process mainly includes the incorporation of DNA labeled with cholesterol and thiol groups into EV membranes and subsequent deposition of Au3+ and Pt4+ to allow the DNA-functionalized EVs to be encapsulated with AuPt nanoshells. It is found that the synthesized AuPt-metallized EVs possess extrinsic peroxidase-like activity. Utilizing the feature of the catalytic metal nanoshells just growth on the EV membranes, the DEVM method enables multiparametric recognition of target proteins and EV membranes and can produce an amplified colorimetric signal, avoiding the interference of free proteins. By profiling four surface proteins of EVs from 48 patients with pulmonary nodules, the highest area under the receiver operating characteristic curve (0.9983) is obtained. Therefore, this work provides a feasible EVs analysis tool for accurate pulmonary nodules management.


Asunto(s)
Vesículas Extracelulares , Proteínas de la Membrana , Humanos , Biomarcadores/metabolismo , Proteínas de la Membrana/metabolismo , ADN/metabolismo , Vesículas Extracelulares/metabolismo
9.
Mol Ther ; 30(8): 2817-2827, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35450820

RESUMEN

Chimeric antigen receptor T (CAR-T) cell therapy has faced a series of challenges and has shown very little efficacy in solid tumors to date. Although genetically engineered macrophages have achieved definite therapeutic effect in solid tumors, heterogeneous expression of engineered proteins and the potential for toxicity limit further applications. Herein, we propose a nongenetic and simple macrophage cell engineering strategy through glycan metabolic labeling and click reaction for the treatment of solid tumors. The aptamer-engineered M1 macrophage (ApEn-M1) showed enhanced active targeting ability for tumor cells in vitro and in vivo, resulting in significant cytotoxicity effects. Moreover, ApEn-M1 exhibited superior antitumor efficacy in a breast cancer xenograft mouse model and a lung metastasis mouse model of breast cancer. Interestingly, the ApEn-M1 could reprogram the immunity microenvironment by increasing T cell infiltration and enhancing T cell activity in the tumor region. Additionally, the administration of ApEn-M1 showed no obvious systemic side effects. With glycan metabolic labeling, the macrophages could be efficiently labeled with aptamers on the cell surface via click reaction without genetic alteration or cell damage. Hence, this study serves as a proof of concept for cell-surface anchor engineering and expands the range of nongenetic macrophage cell engineering strategies.


Asunto(s)
Neoplasias Pulmonares , Neoplasias , Animales , Línea Celular Tumoral , Humanos , Inmunoterapia/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Ratones , Neoplasias/patología , Linfocitos T , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Nanobiotechnology ; 21(1): 234, 2023 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-37481551

RESUMEN

The rapid and accurate identification of methicillin-resistant Staphylococcus aureus at an early antibiotic therapy stage would be benefit to disease diagnosis and antibiotic selection. Herein, we integrated cross-priming amplification (CPA) and CRISPR/Cas 12a (designated as CPA-Cas 12a) systems to establish a sensitive and efficient lateral flow assay to detect methicillin-resistant Staphylococcus aureus. This assay relies on the CPA isothermal nucleic acid amplification strategy which can amplify the DNA extracted from Staphylococcus aureus and accompanying the indiscriminately trans-cleavage process of Cas 12a/CrRNA duplex after recognizing specific sequence. Taking the advantage of reporter and high turnover Cas 12a activity, a dramatic change in response was achieved to produce a significant increase in the analytical sensitivity. The signal conversion and output were realized using a lateral flow strip to achieve field-deployable detection. Furthermore, this bioassay was accommodated with a microfluidic device to realize automatically portable detection. This proposed assay completed within 30 min with the detection limit of 5 CFU mL-1, was verified by testing bacterial suspension and 202 clinical samples. Given the high sensitivity, specificity and efficiency, this colorimetric readout assay through strip could be further promoted to the clinical diagnosis, clinical medication of multidrug-resistant bacteria.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Sistemas CRISPR-Cas , Reactividad Cruzada , Staphylococcus aureus , Antibacterianos/farmacología , Bioensayo
11.
J Nanobiotechnology ; 21(1): 86, 2023 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-36906540

RESUMEN

Programmed cell death ligand 1 protein-positive (PD-L1+) exosomes have been found to be a potential biomarker for the diagnosis of non-small cell lung cancer (NSCLC). However, the development of highly sensitive detection technique for PD-L1+ exosomes is still a challenge in clinical applications. Herein, a sandwich electrochemical aptasensor based on ternary metal-metalloid palladium-copper-boron alloy microporous nanospheres (PdCuB MNs) and Au@CuCl2 nanowires (NWs) was designed for the detection of PD-L1+ exosomes. The excellent peroxidase-like catalytic activity of PdCuB MNs and the high conductivity of Au@CuCl2 NWs endow the fabricated aptasensor with intense electrochemical signal, thus enabling the detection of low abundance exosomes. The analytical results revealed that the aptasensor maintained favorable linearity over a wide concentration range of 6 orders of magnitude and reached a low detection limit of 36 particles/mL. The aptasensor is successfully applied to the analysis of complex serum samples and achieves the accurate identification of clinical NSCLC patients. Overall, the developed electrochemical aptasensor provides a powerful tool for early diagnosis of NSCLC.


Asunto(s)
Aptámeros de Nucleótidos , Técnicas Biosensibles , Carcinoma de Pulmón de Células no Pequeñas , Exosomas , Neoplasias Pulmonares , Nanopartículas del Metal , Nanocables , Humanos , Antígeno B7-H1 , Técnicas Biosensibles/métodos , Límite de Detección , Oro
12.
J Nanobiotechnology ; 21(1): 122, 2023 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-37031177

RESUMEN

How to achieve delicate regulation of enzyme activity and empower it with more roles is the peak in the field of enzyme catalysis research. Traditional proteases or novel nano-enzymes are unable to achieve stimulus-responsive activity modulation due to their own structural limitations. Here, we propose a novel Controllable Enzyme Activity Switch, CEAS, based on hemin aggregation regulation, to deeply explore its regulatory mechanism and develop multimodal biosensing applications. The core of CEAS relies on the dimerizable inactivation of catalytically active center hemin and utilizes a DNA template to orderly guide the G4-Hemin DNAzyme to tightly bind to DNA-Hemin, thereby shutting down the catalytic ability. By customizing the design of the guide template, different target stimulus responses lead to hemin dimerization dissociation and restore the synergistic catalysis of G4-Hemin and DNA-Hemin, thus achieving a target-regulated enzymatic activity switch. Moreover, the programmability of CEAS allowed it easy to couple with a variety of DNA recognition and amplification techniques, thus developing a series of visual protein detection systems and highly sensitive fluorescent detection systems with excellent bioanalytical performance. Therefore, the construction of CEAS is expected to break the limitation of conventional enzymes that cannot be targetable regulated, thus enabling customizable enzymatic reaction systems and providing a new paradigm for controllable enzyme activities.


Asunto(s)
Técnicas Biosensibles , ADN Catalítico , G-Cuádruplex , Hemina/química , Técnicas Biosensibles/métodos , ADN , ADN Catalítico/química , ADN Catalítico/genética , ADN Catalítico/metabolismo
13.
Nucleic Acids Res ; 49(22): 13031-13044, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34878146

RESUMEN

G-quadruplex (G4)/hemin DNAzyme is promising horseradish peroxidase (HRP)-mimic candidate in the biological field. However, its relatively unsatisfactory catalytic capacity limits the potential applications. Inspired by nature protease, we conducted a proximity-enhanced cofactor assembly strategy (PECA) to form an exceptional HRP mimic, namely zippered G4/hemin DNAzyme (Z-G4/H). The hybridization of short oligonucleotides induced proximity assembly of the DNA-grafted hemin (DGH) with the complementary G4 sequences (cG4s), mimicking the tight configuration of protease cofactor and apoenzyme. The detailed investigations of catalytic efficiency and mechanism verified the higher activity, more rapid catalytic rate and high environmental tolerance of the Z-G4/H than the classical G4/hemin DNAzymes (C-G4/H). Furthermore, a proximity recognition transducer has been developed based on the PECA for sensitive detection of gene rearrangement and imaging human epidermal growth factor receptor 2 protein (HER2) dimerization on cell surfaces. Our studies demonstrate the high efficiency of Z-G4/H and its universal application potential in clinical diagnostics and biomolecule interaction research. It also may offer significant opportunities and inspiration for the engineering of the protease-free mimic enzyme.


Asunto(s)
ADN Catalítico/metabolismo , Pruebas de Enzimas/métodos , G-Cuádruplex , Hemina/metabolismo , Biocatálisis , Línea Celular Tumoral , Dicroismo Circular/métodos , ADN Catalítico/genética , Estabilidad de Enzimas , Hemina/química , Humanos , Concentración de Iones de Hidrógeno , Cinética , Células MCF-7 , Estructura Molecular , Mutación , Espectrofotometría/métodos , Temperatura
14.
J Nanobiotechnology ; 20(1): 357, 2022 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-35918755

RESUMEN

The exosomal miRNA (exo-miRNA) derived from tumor cells contains rich biological information that can effectively aid in the early diagnosis of disease. However, the extremely low abundance imposes stringent requirements for accurate detection techniques. In this study, a novel, protease-free DNA amplification strategy, known as "Rolling Hoop Orbital Amplification" (RHOA), was initially developed based on the design concept of local reaction and inspired by the childhood game of rolling iron ring. Benefiting from the local space constructed by the DNA orbital, the circular DNA enzyme rolls directionally and interacts efficiently with the amplification element, making it nearly 3-fold more productive than conventional free-diffusion amplification. Similarly, the localized cascade nanozyme catalytic system formed by bridging DNA probes also exhibits outperformed than free ones. Therefore, a localized energized high-performance electrochemiluminescence (ECL) biosensor was constructed by bridging cascading nanozymes on the electrode surface through DNA probes generated by RHOA, with an impressive limit of detection (LOD) of 1.5 aM for the detection of exosomal miRNA15a-5p and a stable linearity over a wide concentration range from 10- 2 to 108 fM. Thus, this work is a focused attempt at the localized reaction, which is expected to provide a reliable method for accurately detecting of exo-miRNAs.


Asunto(s)
Técnicas Biosensibles , MicroARNs , Técnicas Biosensibles/métodos , ADN/genética , Sondas de ADN , Límite de Detección , MicroARNs/genética , Técnicas de Amplificación de Ácido Nucleico/métodos
15.
Mikrochim Acta ; 189(9): 355, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36038693

RESUMEN

A multifunctional catalytic nanomaterial (Co-MOF@AuNP@ABEI) composed of cobalt-doped metal-organic frameworks (Co-MOF), gold nanoparticles (AuNP), and N-(4-aminobutyl)-N-(ethylisoluminol) (ABEI) is reported. Co-MOF@AuNP@ABEI exhibits high synergistic and zero-distance catalytic properties, which are beneficial to the improvement of the detection sensitivity of an electrochemiluminescent (ECL) biosensor. After coupling with the ECL system and 3D magnetic walking nanomachine amplification strategy, the Co-MOF@AuNP@ABEI can achieve an ultrasensitive ECL assay of Burkholderia pseudomallei with the limit of detection (LOD) of 60.3 aM, which is 2 and 4 orders of magnitude lower than individual ECL system without the nanomachine (4.97 fM) and individual walking nanomachine (340 fM), and superior to the pathogenic bacteria analyses in the previous report. Moreover, the LOD of the proposed ECL detection system for the determination of B. pseudomallei in serum sample was as low as 9.0 CFU mL-1. The relative standard deviations (RSD) of ECL intensity for the detection of five B. pseudomallei-spiked serum samples were 4.02%, 0.84%, 0.84%, 1.55%, and 0.21%, respectively. The recoveries of the ECL biosensor for the detection of B. pseudomallei DNA-spiked serum samples were 93.63 ~ 107.83%. Therefore, this work demonstrated that the developed multifunctional catalytic nanomaterial with synergistic and zero-distance catalytic properties can be used as excellent ECL signal reporter to improve the detection sensitivity of ECL biosensor.


Asunto(s)
Técnicas Biosensibles , Burkholderia pseudomallei , Luminol/análogos & derivados , Nanopartículas del Metal , Estructuras Metalorgánicas , Cobalto , Técnicas Electroquímicas , Oro , Mediciones Luminiscentes , Luminol/química
16.
J Nanobiotechnology ; 19(1): 225, 2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34325706

RESUMEN

BACKGROUND: Anti-myeloperoxidase antibody (anti-MPO) is an important biomarker for anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitides (AAVs). However, the complicated operation procedures and insufficient sensitivity of conventional anti-MPO detection methods limit their application in monitoring efficacy of AAVs in clinical diagnosis. Herein, a dual amplified electrochemiluminescence (ECL) immunosensor based on multi-function PtCo nanozymes/CdS nanocrystals@graphene oxide (PtCo/CdS@GO) luminophores and K2S2O8/H2O2 coreactants has been fabricated for ultrasensitive detection of anti-MPO. RESULTS: PtCo/CdS@GO luminophores as novel signal amplification labels and nanocarriers to load rabbit anti-mouse IgG were synthesized by co-doping with Pt and Co nanozymes simultaneously with several considerable advantages, including astonishing peroxidase-like catalytic activity, high-efficiency luminescence performance and superior stability in aqueous solutions. Meanwhile, upon the K2S2O8/H2O2 coreactants system, benefiting from the efficient peroxidase-like activity of the PtCo/CdS@GO toward H2O2, massive of transient reactive intermediates could react with K2S2O8, thus obtaining higher ECL emission. Therefore, the developed ECL immunosensor for anti-MPO detection displayed good analytical performance with good concentration linearity in the range of 0.02 to 1000 pg/mL and low detection limit down to 7.39 fg/mL. CONCLUSIONS: The introduction of multi-function PtCo/CdS@GO luminophores into the established ECL immunoassay not only was successfully applied for specific detection of anti-MPO in clinical serum samples, but also provided a completely new concept to design other high-performance luminophores. Meaningfully, the ECL immunoassay strategy held wide potential for biomarkers detection in clinical diagnosis.


Asunto(s)
Técnicas Biosensibles/métodos , Grafito/metabolismo , Peróxido de Hidrógeno/metabolismo , Peroxidasa/inmunología , Animales , Inmunoensayo/métodos , Luminiscencia , Nanopartículas del Metal , Ratones , Nanopartículas , Conejos
17.
J Nanobiotechnology ; 19(1): 272, 2021 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-34496877

RESUMEN

BACKGROUND: Retinol binding protein 4 (RBP4) has been regarded as an important serological biomarker for type 2 diabetes mellitus (T2DM). Hence, the construction of a highly sensitive detection method for RBP4 is the key to early prevention and multidisciplinary intervention of T2DM. In this work, a dual-quenched electrochemiluminescence (ECL) immunosensor has been fabricated for ultrasensitive detection of RBP4 by combining zeolitic imidazolate framework-67/AuPt-supported luminol (luminol@AuPt/ZIF-67) with MnO2 nanosheets-grown on carbon nanotubes (MnO2@CNTs). RESULTS: AuPt/ZIF-67 hybrids with high-efficiency peroxidase-like activity could provide multipoint binding sites for luminol and antibodies and significantly boost the amplified initial signal of the ECL immunosensor. Upon glutathione/H2O2 coreactants system, MnO2@CNTs composites could quench the initial signal by inhibiting mimic peroxidase activity of luminol@AuPt/ZIF-67. Moreover, the absorption spectrum of the MnO2@CNTs composites completely overlaps with the emission spectrum of luminol, which can further reduce initial signal by ECL resonance energy transfer (ECL-RET). CONCLUSIONS: Benefiting from the above-mentioned properties, the designed immunoassay sensitivity exhibited excellent sensitivity and relative stability for RBP4 detection range from 0.0001 to 100 ng mL-1 with a low detection limit of 43 fg mL-1. Therefore, our ECL immunosensor provides an alternative assaying strategy for early diagnosis of T2DM.


Asunto(s)
Inmunoensayo/métodos , Luminol/química , Estructuras Metalorgánicas/química , Nanocompuestos/química , Proteínas Plasmáticas de Unión al Retinol/análisis , Técnicas Electroquímicas , Oro/química , Humanos , Límite de Detección , Mediciones Luminiscentes , Compuestos de Manganeso/química , Nanotubos de Carbono/química , Óxidos/química , Platino (Metal)/química , Reproducibilidad de los Resultados
18.
J Nanobiotechnology ; 19(1): 408, 2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34876148

RESUMEN

In this study, a novel electrochemical biosensor was constructed for ultrasensitive and locus-specific detection of N6-Methyladenine (m6A) in DNA using double-hindered replication and nucleic acid-coated methylene blue (MB)@Zr-MOF. Based on the combination of m6A-impeded replication and AgI-mediated mismatch replication, this mode could effectively stop the extension of the strand once DNA polymerase encountered m6A site, which specifically distinguish the m6A site from natural A site in DNA. Also, Zr-MOF with high porosity and negative surface potential features was carefully chose to load cationic MB, resulting a stable and robust MB@Zr-MOF electrochemical tag. As a result, the developed biosensor exhibited a wide linear range from 1 fM to 1 nM with detection limit down to 0.89 fM. Profiting from the high sensitivity and selectivity, the biosensing strategy revealed good applicability, which had been demonstrated by quantitating m6A DNA at specific site in biological matrix. Thus, the biosensor provides a promising platform for locus-specific m6A DNA analysis.


Asunto(s)
Adenina/análogos & derivados , Técnicas Biosensibles/métodos , ADN/química , Estructuras Metalorgánicas/química , Azul de Metileno/química , Adenina/análisis , Adenina/química , Límite de Detección , Nanotecnología , Análisis de Secuencia de ADN , Circonio/química
19.
J Nanobiotechnology ; 19(1): 450, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34952586

RESUMEN

Human epidermal growth factor receptor 2 (HER2)-positive exosomes play an extremely important role in the diagnosis and treatment options of breast cancers. Herein, based on the reformative tyramine signal amplification (TSA) enabled by molecular aptamer beacon (MAB) conversion, a label-free surface plasmon resonance (SPR) biosensor was proposed for highly sensitive and specific detection of HER2-positive exosomes. The exosomes were captured by the HER2 aptamer region of MAB immobilized on the chip surface, which enabled the exposure of the G-quadruplex DNA (G4 DNA) that could form peroxidase-like G4-hemin. In turn, the formed G4-hemin catalyzed the deposition of plentiful tyramine-coated gold nanoparticles (AuNPs-Ty) on the exosome membrane with the help of H2O2, generating a significantly enhanced SPR signal. In the reformative TSA system, the horseradish peroxidase (HRP) as a major component was replaced with nonenzymic G4-hemin, bypassing the defects of natural enzymes. Moreover, the dual-recognition of the surface proteins and lipid membrane of the desired exosomes endowed the sensing strategy with high specificity without the interruption of free proteins. As a result, this developed SPR biosensor exhibited a wide linear range from 1.0 × 104 to 1.0 × 107 particles/mL. Importantly, this strategy was able to accurately distinguish HER2-positive breast cancer patients from healthy individuals, exhibiting great potential clinical application.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Exosomas/metabolismo , Resonancia por Plasmón de Superficie , Tiramina/química , Neoplasias de la Mama/diagnóstico , Línea Celular Tumoral , Exosomas/química , G-Cuádruplex , Oro/química , Hemina/química , Humanos , Peróxido de Hidrógeno/química , Límite de Detección , Nanopartículas del Metal/química , Receptor ErbB-2/metabolismo , Reproducibilidad de los Resultados
20.
Mikrochim Acta ; 188(10): 319, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34476628

RESUMEN

A novel cytosensor was constructed for the ultrasensitive detection and nondestructive release of circulating tumor cells (CTCs) by combining Au nanoparticles-loaded two-dimensional bimetallic PdMo (2D Au@PdMo) nanozymes and electrochemical reductive desorption. The 2D Au@PdMo nanozymes possessed high-efficiency peroxidase-like activity and were assembled with an aptamer composed of a thiol-modified epithelial specific cell adhesion molecule (EpCAM) to strengthen CTCs adhesion. Moreover, the electrode surface was decorated with highly fractal Au nanostructures (HFAuNSs) composites due to the similarity in fractal nanostructure with the CTCs membrane to enhance the CTCs anchoring efficiency and release capability. The captured CTCs could be further efficiently dissociated and nondestructively released from the modified electrodes upon electrochemical reductive desorption. The designed cytosensor showed an excellent analytical performance, with a wide linear range from 2 to 1 × 105 cells mL-1 and low limit of detection (LOD) of 2 cells mL-1 (S/N = 3) at the working potential in the range  -0.6 to 0.2 V. A satisfactory CTCs release reaching a range of 93.7-97.4% with acceptable RSD from 3.55 to 6.41% and good cell viability was obtained. Thus, the developed cytosensor might provide a potential alternative to perform CTC-based liquid biopsies, with promising applications in early diagnosis of tumors. Preparation and mechanism of desorption of the cytosensor based on 2D Au@PdMo nanozymes and electrochemical reductive desorption for the detection and release of CTCs. A Preparation procedure of the Apt/Au@PbMo bioconjugates. B Fabrication process of the sandwich-type cytosensor. C Electrochemical signal produced by the Au@PdMo nanozymes. D Mechanism of electrochemical reductive desorption for CTCs release.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Separación Celular/métodos , Nanopartículas del Metal/química , Células Neoplásicas Circulantes/química , Aptámeros de Nucleótidos/química , Catálisis , Línea Celular Tumoral , Técnicas Electroquímicas/métodos , Molécula de Adhesión Celular Epitelial/metabolismo , Oro/química , Humanos , Límite de Detección , Molibdeno/química , Paladio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA