Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 10(31): e2303285, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37587020

RESUMEN

The extensive and improper use of antibiotics has led to a dramatic increase in the frequency of antibiotic resistance among human pathogens, complicating infectious disease treatments. In this work, a method for rapid antimicrobial susceptibility testing (AST) is presented using microstructured silicon diffraction gratings integrated into prototype devices, which enhance bacteria-surface interactions and promote bacterial colonization. The silicon microstructures act also as optical sensors for monitoring bacterial growth upon exposure to antibiotics in a real-time and label-free manner via intensity-based phase-shift reflectometric interference spectroscopic measurements (iPRISM). Rapid AST using clinical isolates of Escherichia coli (E. coli) from urine is established and the assay is applied directly on unprocessed urine samples from urinary tract infection patients. When coupled with a machine learning algorithm trained on clinical samples, the iPRISM AST is able to predict the resistance or susceptibility of a new clinical sample with an Area Under the Receiver Operating Characteristic curve (AUC) of ∼ 0.85 in 1 h, and AUC > 0.9 in 90 min, when compared to state-of-the-art automated AST methods used in the clinic while being an order of magnitude faster.


Asunto(s)
Escherichia coli , Silicio , Humanos , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Pruebas en el Punto de Atención
2.
Nanoscale Horiz ; 7(7): 729-742, 2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35616534

RESUMEN

With new advances in infectious disease, antifouling surfaces, and environmental microbiology research comes the need to understand and control the accumulation and attachment of bacterial cells on a surface. Thus, we employ intrinsic phase-shift reflectometric interference spectroscopic measurements of silicon diffraction gratings to non-destructively observe the interactions between bacterial cells and abiotic, microstructured surfaces in a label-free and real-time manner. We conclude that the combination of specific material characteristics (i.e., substrate surface charge and topology) and characteristics of the bacterial cells (i.e., motility, cell charge, biofilm formation, and physiology) drive bacteria to adhere to a particular surface, often leading to a biofilm formation. Such knowledge can be exploited to predict antibiotic efficacy and biofilm formation, and enhance surface-based biosensor development, as well as the design of anti-biofouling strategies.


Asunto(s)
Adhesión Bacteriana , Incrustaciones Biológicas , Bacterias , Incrustaciones Biológicas/prevención & control , Microbiología Ambiental , Silicio
3.
Eng Life Sci ; 22(3-4): 319-333, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35382545

RESUMEN

Since the invention of the first biosensors 70 years ago, they have turned into valuable and versatile tools for various applications, ranging from disease diagnosis to environmental monitoring. Traditionally, antibodies have been employed as the capture probes in most biosensors, owing to their innate ability to bind their target with high affinity and specificity, and are still considered as the gold standard. Yet, the resulting immunosensors often suffer from considerable limitations, which are mainly ascribed to the antibody size, conjugation chemistry, stability, and costs. Over the past decade, aptamers have emerged as promising alternative capture probes presenting some advantages over existing constraints of immunosensors, as well as new biosensing concepts. Herein, we review the employment of antibodies and aptamers as capture probes in biosensing platforms, addressing the main aspects of biosensor design and mechanism. We also aim to compare both capture probe classes from theoretical and experimental perspectives. Yet, we highlight that such comparisons are not straightforward, and these two families of capture probes should not be necessarily perceived as competing but rather as complementary. We, thus, elaborate on their combined use in hybrid biosensing schemes benefiting from the advantages of each biorecognition element.

4.
Adv Biochem Eng Biotechnol ; 179: 247-265, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-32435872

RESUMEN

The recent coronavirus (COVID-19) pandemic has underscored the need to move from traditional lab-centralized diagnostics to point-of-care (PoC) settings. Lab-on-a-chip (LoC) platforms facilitate the translation to PoC settings via the miniaturization, portability, integration, and automation of multiple assay functions onto a single chip. For this purpose, paper-based assays and microfluidic platforms are currently being extensively studied, and much focus is being directed towards simplifying their design while simultaneously improving multiplexing and automation capabilities. Signal amplification strategies are being applied to improve the performance of assays with respect to both sensitivity and selectivity, while smartphones are being integrated to expand the analytical power of the technology and promote its accessibility. In this chapter, we review the main technologies in the field of LoC platforms for PoC medical diagnostics and survey recent approaches for improving these assays.


Asunto(s)
COVID-19 , Técnicas Analíticas Microfluídicas , COVID-19/diagnóstico , Humanos , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/métodos , Microfluídica , Sistemas de Atención de Punto , Teléfono Inteligente
5.
Talanta ; 239: 123124, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896821

RESUMEN

We present a porous Si (PSi)-based label-free optical biosensor for sensitive and continuous detection of a model target protein biomarker in gastrointestinal (GI) tract fluids. The biosensing platform is designed to continuously monitor its target protein within the complex GI fluids without sample preparation and washing steps. An oxidized PSi Fabry-Pérot thin films are functionalized with aptamers, which are used as the capture probes. The optical response of the aptamer-conjugated PSi is studied upon exposure to unprocessed GI fluids, originated from domestic pigs, spiked with the target protein. We investigate biological and chemical surface passivation methods to stabilize the surface and reduce non-specific adsorption of interfering proteins and molecules within the GI fluids. For the passivated PSi aptasensor we simulate continuous in vivo biosensing conditions, demonstrating that the aptasensor could successfully detect the target in a continuous manner without any need for surface washing after the target protein binding events, at a clinically relevant range. Furthermore, we simulate biosensing conditions within a smart capsule, in which the aptasensor is occasionally exposed to GI fluids in flow or via repeated cycles of injection and static incubation events. Such biosensor can be implemented within ingestible capsule devices and used for in situ biomarker detection in the GI tract.


Asunto(s)
Aptámeros de Nucleótidos , Técnicas Biosensibles , Biomarcadores , Tracto Gastrointestinal , Silicio
6.
ACS Meas Sci Au ; 1(2): 82-94, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34693403

RESUMEN

The anterior gradient homologue-2 (AGR2) protein is an attractive biomarker for various types of cancer. In pancreatic cancer, it is secreted to the pancreatic juice by premalignant lesions, which would be an ideal stage for diagnosis. Thus, designing assays for the sensitive detection of AGR2 would be highly valuable for the potential early diagnosis of pancreatic and other types of cancer. Herein, we present a biosensor for label-free AGR2 detection and investigate approaches for enhancing the aptasensor sensitivity by accelerating the target mass transfer rate and reducing the system noise. The biosensor is based on a nanostructured porous silicon thin film that is decorated with anti-AGR2 aptamers, where real-time monitoring of the reflectance changes enables the detection and quantification of AGR2, as well as the study of the diffusion and target-aptamer binding kinetics. The aptasensor is highly selective for AGR2 and can detect the protein in simulated pancreatic juice, where its concentration is outnumbered by orders of magnitude by numerous proteins. The aptasensor's analytical performance is characterized with a linear detection range of 0.05-2 mg mL-1, an apparent dissociation constant of 21 ± 1 µM, and a limit of detection of 9.2 µg mL-1 (0.2 µM), which is attributed to mass transfer limitations. To improve the latter, we applied different strategies to increase the diffusion flux to and within the nanostructure, such as the application of isotachophoresis for the preconcentration of AGR2 on the aptasensor, mixing, or integration with microchannels. By combining these approaches with a new signal processing technique that employs Morlet wavelet filtering and phase analysis, we achieve a limit of detection of 15 nM without compromising the biosensor's selectivity and specificity.

7.
ACS Sens ; 6(8): 2967-2978, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34387077

RESUMEN

The ultimate detection limit of optical biosensors is often limited by various noise sources, including those introduced by the optical measurement setup. While sophisticated modifications to instrumentation may reduce noise, a simpler approach that can benefit all sensor platforms is the application of signal processing to minimize the deleterious effects of noise. In this work, we show that applying complex Morlet wavelet convolution to Fabry-Pérot interference fringes characteristic of thin film reflectometric biosensors effectively filters out white noise and low-frequency reflectance variations. Subsequent calculation of the average difference in extracted phase between the filtered analyte and reference signals enables a significant reduction in the limit of detection (LOD). This method is applied on experimental data sets of thin film porous silicon sensors (PSi) in buffered solution and complex media obtained from two different laboratories. The demonstrated improvement in the LOD achieved using wavelet convolution and average phase difference paves the way for PSi optical biosensors to operate with clinically relevant detection limits for medical diagnostics, environmental monitoring, and food safety.


Asunto(s)
Técnicas Biosensibles , Límite de Detección , Porosidad , Silicio
8.
Mikrochim Acta ; 188(3): 67, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33543321

RESUMEN

Microfluidic integration of biosensors enables improved biosensing performance and sophisticated lab-on-a-chip platform design for numerous applications. While soft lithography and polydimethylsiloxane (PDMS)-based microfluidics are still considered the gold standard, 3D-printing has emerged as a promising fabrication alternative for microfluidic systems. Herein, a 3D-printed polyacrylate-based microfluidic platform is integrated for the first time with a label-free porous silicon (PSi)-based optical aptasensor via a facile bonding method. The latter utilizes a UV-curable adhesive as an intermediate layer, while preserving the delicate nanostructure of the porous regions within the microchannels. As a proof-of-concept, a generic model aptasensor for label-free detection of his-tagged proteins is constructed, characterized, and compared to non-microfluidic and PDMS-based microfluidic setups. Detection of the target protein is carried out by real-time monitoring reflectivity changes of the PSi, induced by the target binding to the immobilized aptamers within the porous nanostructure. The microfluidic integrated aptasensor has been successfully used for detection of a model target protein, in the range 0.25 to 18 µM, with a good selectivity and an improved limit of detection, when compared to a non-microfluidic biosensing platform (0.04 µM vs. 2.7 µM, respectively). Furthermore, a superior performance of the 3D-printed microfluidic aptasensor is obtained, compared to a conventional PDMS-based microfluidic platform with similar dimensions.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Glicósido Hidrolasas/análisis , Técnicas Analíticas Microfluídicas/métodos , Resinas Acrílicas/química , Glicósido Hidrolasas/química , Ácidos Nucleicos Inmovilizados/química , Dispositivos Laboratorio en un Chip , Límite de Detección , Técnicas Analíticas Microfluídicas/instrumentación , Porosidad , Impresión Tridimensional , Prueba de Estudio Conceptual , Silicio/química
9.
ACS Sens ; 5(10): 3058-3069, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32896130

RESUMEN

Porous silicon (PSi) thin films have been widely studied for biosensing applications, enabling label-free optical detection of numerous targets. The large surface area of these biosensors has been commonly recognized as one of the main advantages of the PSi nanostructure. However, in practice, without application of signal amplification strategies, PSi-based biosensors suffer from limited sensitivity, compared to planar counterparts. Using a theoretical model, which describes the complex mass transport phenomena and reaction kinetics in these porous nanomaterials, we reveal that the interrelated effect of bulk and hindered diffusion is the main limiting factor of PSi-based biosensors. Thus, without significantly accelerating the mass transport to and within the nanostructure, the target capture performance of these biosensors would be comparable, regardless of the nature of the capture probe-target pair. We use our model to investigate the effect of various structural and biosensor characteristics on the capture performance of such biosensors and suggest rules of thumb for their optimization.


Asunto(s)
Técnicas Biosensibles , Nanoestructuras , Porosidad , Silicio
10.
Analyst ; 145(14): 4991-5003, 2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32519701

RESUMEN

Over the past decade aptamers have emerged as a promising class of bioreceptors for biosensing applications with significant advantages over conventional antibodies. However, experimental studies comparing aptasensors and immunosensors, under equivalent conditions, are limited and the results are inconclusive, in terms of benefits and limitations of each bioreceptor type. In the present work, the performance of aptamer and antibody bioreceptors for the detection of a his-tagged protein, used as a model target, is compared. The bioreceptors are immobilized onto a nanostructured porous silicon (PSi) thin film, used as the optical transducer, and the target protein is detected in a real-time and label-free format by reflective interferometric Fourier transform spectroscopy. For the antibodies, random-oriented immobilization onto the PSi nanostructure results in a poor biosensing performance. Contrary, Fc-oriented immobilization of the antibodies shows a similar biosensing performance to that exhibited by the aptamer-based biosensor, in terms of binding rate, dynamic detection range, limit of detection and selectivity. The aptasensor outperforms in terms of its reusability and storability, while the immunosensor could not be regenerated for subsequent experiments.


Asunto(s)
Aptámeros de Nucleótidos , Técnicas Biosensibles , Inmunoensayo , Porosidad , Silicio
12.
ACS Sens ; 2(12): 1767-1773, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29164872

RESUMEN

Porous silicon (PSi) nanomaterials have been widely studied as label-free optical biosensors for protein detection. However, these biosensors' performance, specifically in terms of their sensitivity (which is typically in the micromolar range), is insufficient for many applications. Herein, we present a proof-of-concept application of the electrokinetic isotachophoresis (ITP) technique for real-time preconcentration of a target protein on a PSi biosensor. With ITP, a highly concentrated target zone is delivered to the sensing area, where the protein target is captured by immobilized aptamers. The detection of the binding events is conducted in a label-free manner by reflective interferometric Fourier transformation spectroscopy (RIFTS). Up to 1000-fold enhancement in local concentration of the protein target and the biosensor's sensitivity are achieved, with a measured limit of detection of 7.5 nM. Furthermore, the assay is successfully performed in complex media, such as bacteria lysate samples, while the selectivity of the biosensor is retained. The presented assay could be further utilized for other protein targets, and to promote the development of clinically useful PSi biosensors.


Asunto(s)
Técnicas Biosensibles/métodos , Glicósido Hidrolasas/análisis , Fragmentos de Péptidos/análisis , Silicio/química , Aptámeros de Nucleótidos/química , Técnicas Biosensibles/instrumentación , Escherichia coli K12 , Glicósido Hidrolasas/química , Isotacoforesis/métodos , Dispositivos Laboratorio en un Chip , Límite de Detección , Fragmentos de Péptidos/química , Porosidad , Dominios Proteicos , Sensibilidad y Especificidad , Espectrofotometría/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA