Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Langmuir ; 40(27): 13957-13967, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38919992

RESUMEN

The specificity and efficiency of enzyme-mediated reactions have the potential to positively impact many biotechnologies; however, many enzymes are easily degraded. Immobilization on a solid support has recently been explored to improve enzyme stability. This study aims to gain insights and facilitate enzyme adsorption onto gold nanoparticles (AuNPs) to form a stable bioconjugate through the installation of thiol functional groups that alter the protein chemistry. In specific, the model enzyme, horseradish peroxidase (HRP), is thiolated via Traut's reagent to increase the robustness and enzymatic activity of the bioconjugate. This study compares HRP and its thiolated analog (THRP) to deduce the impact of thiolation and AuNP-immobilization on the enzyme activity and stability. HRP, THRP, and their corresponding bioconjugates, HRP-AuNP and THRP-AuNP, were analyzed via UV-vis spectrophotometry, circular dichroism, zeta potential, and enzyme-substrate kinetics assays. Our data show a 5-fold greater adsorption for THRP on the AuNP, in comparison to HRP, that translated to a 5-fold increase in the THRP-AuNP bioconjugate activity. The thiolated and immobilized HRP exhibited a substantial improvement in stability at elevated temperatures (50 °C) and storage times (1 month) relative to the native enzyme in solution. Moreover, HRP, THRP, and their bioconjugates were incubated with trypsin to assess the susceptibility to proteolytic digestion. Our results demonstrate that THRP-AuNP bioconjugates maintain full enzymatic activity after 18 h of incubation with trypsin, whereas free HRP, free THRP, and HRP-AuNP conjugates are rendered inactive by trypsin treatment. These results highlight the potential for protein modification and immobilization to substantially extend enzyme shelf life, resist protease digestion, and enhance biological function to realize enzyme-enabled biotechnologies.


Asunto(s)
Estabilidad de Enzimas , Enzimas Inmovilizadas , Oro , Peroxidasa de Rábano Silvestre , Nanopartículas del Metal , Compuestos de Sulfhidrilo , Oro/química , Peroxidasa de Rábano Silvestre/química , Peroxidasa de Rábano Silvestre/metabolismo , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Compuestos de Sulfhidrilo/química , Nanopartículas del Metal/química , Proteolisis , Adsorción , Cinética
2.
Langmuir ; 39(10): 3720-3728, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36857653

RESUMEN

Proteins adsorbed to gold nanoparticles (AuNPs) form bioconjugates and are critical to many emerging technologies for drug delivery, diagnostics, therapies, and other biomedical applications. A thorough understanding of the interaction between the immobilized protein and AuNP is essential for the bioconjugate to perform as designed. Here, we explore a correlation between the number of solvent-accessible thiol groups on a protein and the protein desorption rate from the AuNP surface in the presence of a competing protein. The chemical modification of human serum albumin (HSA) was carried out to install additional free thiols using Traut's reagent and create a library of HSA analogues by tailoring the molar excess of the Traut's reagent. We pre-adsorbed HSA variants onto the AuNP surface, and the resulting bioconjugates were then exposed to IgG antibody, and protein exchange was monitored as a function of time. We found that the rate of HSA displacement from the AuNP correlated with the experimentally measured number of accessible free thiol groups. Additionally, bioconjugates were synthesized using thiolated analogues of bovine serum albumin (BSA) and suspended in serum as a model for a complex sample matrix. Similarly, desorption rates with serum proteins were modulated with solvent-accessible thiols on the immobilized protein. These results further highlight the key role of Au-S bonds in the formation of protein-AuNP conjugates and provide a pathway to systematically control the number of free thiols on a protein, enabling the controlled release of protein from the surface of AuNP.


Asunto(s)
Nanopartículas del Metal , Albúmina Sérica , Humanos , Oro/química , Nanopartículas del Metal/química , Albúmina Sérica Bovina/química , Albúmina Sérica Humana , Solventes , Compuestos de Sulfhidrilo , Proteínas Inmovilizadas
3.
Analyst ; 149(1): 221-230, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38018888

RESUMEN

Rapid, sensitive, and quantitative detection of biomarkers is needed for early diagnosis of disease and surveillance of infectious outbreaks. Here, we exploit a plasmonic syringe filter and surface-enhanced Raman spectroscopy (SERS) in the development of a rapid detection system, using human IgG as a model diagnostic biomarker. The novel assay design facilitates multiple passages of the sample and labeling solution through the detection zone enabling us to investigate and maximize sampling efficiency to the capture substrate. The vertical flow immunoassay process in this study involves the utilization of filter paper embedded with gold nanoparticles (AuNPs) to form a plasmonic substrate. Capture antibody (anti-human IgG) is then immobilized onto the prepared plasmonic paper and inserted into a vertical flow device (syringe filter holder). Sample solution is passed through the filter paper and the target antigen (human IgG) is selectively captured by the immobilized antibody to form an antibody-antigen complex. Next, functionalized AuNPs as extrinsic Raman labels (ERLs) are passed through the filter paper to label the captured biomarker molecules forming a layered structure. This sandwiched geometry enhances plasmonic coupling and SERS signal to provide highly sensitive detection of biomolecules. Systematic studies to investigate the impact of multiple infuse/withdraw cycles of the sample and labeling solutions reveal that antigen and ERL binding are maximized with 10 and 20 cycles, respectively. The optimized assay achieves a detection limit of ∼0.2 ng mL-1 for human IgG with a total assay time of less than 5 minutes, meeting the demands for rapid point of care diagnostics. Additionally, the optimized platform was implemented in the quantitative analysis of the SARS-CoV-2 nucleocapsid protein, the typical target in commercial, FDA-approved rapid antigen tests for COVID-19.


Asunto(s)
Oro , Nanopartículas del Metal , Humanos , Oro/química , Nanopartículas del Metal/química , Espectrometría Raman/métodos , Biomarcadores , Inmunoensayo/métodos , Antígenos , Inmunoglobulina G
4.
Bioconjug Chem ; 32(8): 1753-1762, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34228917

RESUMEN

Many emerging nanobiotechnologies rely on the proper function of proteins immobilized on gold nanoparticles. Often, the surface chemistry of the AuNP is engineered to control the orientation, surface coverage, and structure of the adsorbed protein to maximize conjugate function. Here, we chemically modified antibody to investigate the effect of protein surface chemistries on adsorption to AuNPs. A monoclonal anti-horseradish peroxidase IgG antibody (anti-HRP) was reacted with N-succinimidyl acrylate (NSA) or reduced dithiobissuccinimidyl propionate (DSP) to modify lysine residues. Zeta potential measurements confirmed that both chemical modifications reduced the localized regions of positive charge on the protein surface, while the DSP modification incorporated additional free thiols. Dynamic light scattering confirmed that native and chemically modified antibodies adsorbed onto AuNPs to form bioconjugates; however, adsorption kinetics revealed that the NSA-modified antibody required significantly more time to allow for the formation of a hard corona. Moreover, conjugates formed with the NSA-modified antibody lost antigen-binding function, whereas unmodified and DSP-modified antibodies adsorbed onto AuNPs to form functional conjugates. These results indicate that high-affinity functional groups are required to prevent protein unfolding and loss of function when adsorbed on the AuNP surface. The reduced protein charge and high-affinity thiol groups on the DSP-modified antibody enabled pH-dependent control of protein orientation and the formation of highly active conjugates at solution pHs (<7.5) that are inaccessible with unmodified antibody due to conjugate aggregation. This study establishes parameters for protein modification to facilitate the formation of highly functional and stable protein-AuNP conjugates.


Asunto(s)
Afinidad de Anticuerpos , Oro/química , Peroxidasa de Rábano Silvestre/inmunología , Inmunoglobulina G/química , Nanopartículas del Metal/química , Acrilatos/química , Adsorción , Concentración de Iones de Hidrógeno , Cinética , Estructura Molecular , Succinimidas/química
5.
Langmuir ; 37(9): 2993-3000, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33621098

RESUMEN

The unique physicochemical properties of gold nanoparticles (AuNPs) provide many opportunities to develop novel biomedical technologies. The surface chemistry of AuNPs can be engineered to perform a variety of functions, including targeted binding, cellular uptake, or stealthlike properties through the immobilization of biomolecules, such as proteins. It is well established that proteins can spontaneously adsorb onto AuNPs, to form a stable and functional bioconjugate; however, the protein-AuNP interaction may result in the formation of less desirable protein-AuNP aggregates. Therefore, it is imperative to investigate the protein-AuNP interaction and elucidate the mechanism by which protein triggers AuNP aggregation. Herein, we systematically investigated the interaction of immunoglobulin G (IgG) antibody with citrate-capped AuNPs as a function of solution pH. We found that the addition of antibody triggers the aggregation of AuNPs for pH < 7.5, whereas a monolayer of antibody adsorbs onto the AuNP to form a stable bioconjugate when the antibody is added to AuNPs at pH ≥ 7.5. Our data identifies electrostatic bridging between the antibody and the negatively charged AuNPs as the mechanism by which aggregation occurs and rules out protein unfolding and surface charge depletion as potential causes. Furthermore, we found that the electrostatic bridging of AuNPs is reversible within the first few hours of interaction, but the protein-AuNP interactions strengthen over 24 h, after which the protein-AuNP aggregate is irreversibly formed. From this data, we developed a straightforward approach to acrylate the basic residues on the antibody to prevent protein-induced aggregation of AuNP over a wide pH range. The results of this study provide additional insight into antibody-nanoparticle interactions and provide a pathway to control the interaction with the potential to enhance the conjugate function.


Asunto(s)
Oro , Nanopartículas del Metal , Anticuerpos
6.
Anal Chem ; 92(9): 6676-6683, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32255335

RESUMEN

Forensic laboratory backlogs are replete with suspected drug samples. Shifting analysis toward the point of seizure would save significant time and public funds. Moreover, a two-tiered identification strategy for controlled substance testing that relies on two independent, discerning methods could entirely circumvent the need for forensic laboratory testing. To this end, we coupled Raman spectroscopy and paper spray ionization mass spectrometry (PSI-MS) on a single instrumental platform. Both methods are capable of ambient analysis with fieldable instruments, yet Raman is often limited to bulk analysis. Critical to this work is the development of a gold nanoparticle (AuNP)-embedded paper swab to extend the capability of Raman spectroscopy to trace evidence via surface-enhanced Raman scattering (SERS). Plasmonic papers are characterized with respect to SERS signals and compatibility with PSI-MS analysis. Proof-of-principle is established with the identification of five representative drugs, and detection limits on the scale of 1-100 ng are achieved for both PSI-MS and SERS. The integrated SERS-PSI-MS system achieved 99.8% accurate chemical identification in a blind study consisting of 500 samples. Additionally, we demonstrate facile discrimination of several JWH-018 isomers via SERS even when MS and MS2 spectra are indistinguishable. Successful coupling of SERS and PSI-MS to enable on-site chemical analysis by two independent methods can potentially lead to a desirable paradigm shift in the handling of drug evidence.

7.
Langmuir ; 36(31): 9241-9249, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32686419

RESUMEN

Protein-gold nanoparticle (AuNP) bioconjugates have many potential applications in nanomedicine. A thorough understanding of the interaction between the protein and the AuNP is critical to engineering these functional bioconjugates with desirable properties. In this work, we investigate the role of free thiols presented by the protein on the stability of the protein-AuNP conjugate. Human serum albumin (HSA) was modified with 2-iminothiolane (Traut's reagent) to introduce additional thiols onto the protein surface, and three variants of HSA were synthesized to present 1, 5, and 20 free thiols by controlling the molar excess of the chemical modifier. Protein exchange studies on AuNPs were conducted using these HSA species and an IgG antibody which exhibited 10 free thiols. Antibody-AuNP conjugates were synthesized, purified, and dispersed in solutions containing each of the HSA species. No protein exchange was detected with the HSA or modified HSA containing 5 thiols; however, 85% of the antibody was displaced on the AuNP surface by the extensively thiolated HSA presenting 20 free thiols. Furthermore, the impact of the protein adsorption sequence was probed in which each of the HSA species were preadsorbed onto the AuNP and dispersed in a solution of antibody. The antibody fully displaced the HSA with a single thiol from the AuNP within 3 h, required 24 h to completely displace the modified HSA containing 5 thiols, and was unable to displace the modified HSA containing 20 thiols. These results indicate that the number of Au-S interactions governs the binding interaction between the protein and the AuNP. This work provides further insight into the protein-AuNP binding mechanism and identifies important design principles for engineered proteins to optimize bioconjugates.


Asunto(s)
Oro , Nanopartículas del Metal , Adsorción , Anticuerpos , Humanos , Compuestos de Sulfhidrilo
8.
Bioconjug Chem ; 30(4): 1182-1191, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30844256

RESUMEN

Novel detection strategies that exploit the unique properties of gold nanoparticles (AuNPs) hold great promise for the advancement of diagnostic testing. Fundamental to many of these nanoparticle-enabled techniques is the immobilization of antibodies onto the AuNP surface to afford selective binding to target species. Orientation and loading density of the immobilized antibodies govern Fab accessibility and are critical to the analytical performance. Here, we use pH to systematically control the surface charge distribution on an antibody and investigate the impact of protein charge on adsorption to AuNPs. Nanoparticle tracking analysis (NTA) is used to measure the adsorption dynamics of anti-horseradish peroxidase antibody (anti-HRP) onto AuNPs at different pHs. NTA enables in situ measurement of antibody adsorption on AuNP by measuring the increase in hydrodynamic diameter ( DH) of the AuNPs as a function of antibody concentration. The adsorption affinity, protein layer thickness, and binding cooperativity at each pH are extracted from the best fit of the adsorption isotherms to the Hill-modified Langmuir equation. Our data show a monolayer of antibody is formed at saturation at pHs 7.5, 8.0, and 8.5, and no difference in anti-HRP-AuNP binding constants is observed in this pH range ( Kd ∼11 nM). However, the increase in DH of the AuNPs with adsorbed protein at monolayer coverage is pH-dependent, measuring 13.2 ± 1.1 nm, 9.8 ± 1.0 nm, and 7.4 ± 0.6 nm for pHs 7.5, 8.0, and 8.5, respectively. Moreover, results of an enzyme-mediated assay reveal the antigen-binding capacity of the immobilized anti-HRP antibody is 33 ± 2%, 23 ± 7%, and 18 ± 2% when adsorbed at pHs 7.5, 8.0, and 8.5, respectively. Our data confirm that antibody charge can be altered with pH to modulate and optimize antibody orientation on AuNP. These studies describe our continued efforts to establish design criteria to prepare conjugates with maximum antigen-binding activity that will enhance the performance of biofunctional nanomaterials.


Asunto(s)
Anticuerpos/química , Oro/química , Concentración de Iones de Hidrógeno , Nanopartículas del Metal/química , Adsorción , Peroxidasa de Rábano Silvestre/química
9.
Langmuir ; 35(32): 10601-10609, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31335148

RESUMEN

Gold nanoparticles (AuNPs) functionalized with proteins to impart desirable surface properties have been developed for many nanobiotechnology applications. A strong interaction between the protein and nanoparticle is critical to the formation of a stable conjugate to realize the potential of these emerging technologies. In this work, we examine the robustness of a protein layer adsorbed onto gold nanoparticles while under the stress of a physiological environment that could potentially lead to protein exchange on the nanoparticle surface. The adsorption interaction of common blood plasma proteins (transferrin, human serum albumin, and fibrinogen) and anti-horseradish peroxidase antibody onto AuNPs is investigated by nanoparticle tracking analysis. Our data show that a monolayer of protein is formed at saturation for each protein, and the maximum size increase for the conjugate, relative to the AuNP core, correlates with the protein size. The binding affinity of each protein to the AuNP is extracted from a best fit of the adsorption isotherm to the Hill equation. The antibody displays the greatest affinity (Kd = 15.2 ± 0.8 nM) that is ∼20-65 times stronger than the affinity of the other plasma proteins. Antibody-AuNP conjugates were prepared, purified, and suspended in solutions of blood plasma proteins to evaluate the stability of the antibody layer. An enzyme-mediated assay confirms that the antibody-AuNP interaction is irreversible, and the adsorbed antibody resists displacement by the plasma proteins. This work provides insight into the capabilities and potential limitations of antibody-AuNP-enabled technologies in biological systems.


Asunto(s)
Anticuerpos/química , Proteínas Sanguíneas/química , Oro/química , Nanopartículas del Metal/química , Adsorción , Animales , Ratones
10.
Bioconjug Chem ; 28(1): 38-42, 2017 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-27689968

RESUMEN

Biomolecular assays based on the aggregation of modified gold nanoparticles (AuNPs) have been developed to provide low detection limits and rapid results with a simple one-step, wash-free procedure. However, a relatively narrow dynamic range, low sensitivity, and poor precision due to time-sensitive readout limit the application of these assay platforms. In this work we synthesized asymmetrically functionalized antibody-AuNP conjugates that are rationally designed to overcome the limitations of aggregation-based immunoassays. Solid-phase synthesis was used to chemically passivate the majority of the AuNP surface and restrict antibody immobilization into a small area of the AuNP surface. These asymmetric conjugates assembled into dimers with the addition of antigen and were stable for over 24 h. In contrast, conventional antibody-AuNP conjugates which are symmetrically modified with antibody assembled into large aggregates that continuously increased in size with the addition of target antigen. These results suggest that asymmetric antibody-AuNP conjugates have the potential to significantly improve the analytical performance of aggregation-based immunoassays.


Asunto(s)
Anticuerpos/química , Oro/química , Inmunoconjugados/química , Nanopartículas del Metal/química , Dimerización , Límite de Detección
11.
Analyst ; 142(23): 4456-4467, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-29091083

RESUMEN

Antibody-modified gold nanoparticles (AuNPs) are central to many novel and emerging biosensing technologies due to the specificity provided by antibody-antigen interactions and the unique properties of nanoparticles. These AuNP-enabled assays have the potential to provide significant improvements in sensitivity and multiplexed analysis compared to conventional immunoassays. However, a major challenge for these AuNP platform technologies is the synthesis of stable antibody-AuNP conjugates that resist aggregation in high salt environments and biological matrices. Moreover, synthetic strategies to form stable conjugates often require different solution conditions, e.g., pH, for each unique antibody. Herein we describe our effort to develop an approach to chemically modify lysine residues on antibodies to facilitate the formation of stable antibody-AuNP conjugates over a wide pH range. In this work, we systematically investigated the immobilization of native and chemically modified antibodies to 60 nm citrate-capped AuNPs as a function of pH and evaluated the stability of the antibody-AuNP conjugate in a saline environment. We have developed a method to chemically modify the lysine residues on an antibody prior to conjugation to the AuNP that results in stable conjugates over a wide pH range (6.0-8.5). Amino acid analysis and zeta potential measurements of native and modified antibodies reveal that the requisite modification correlates with the number of lysine residues, and a reduction in positive charge contribution from protonated lysine is required to form stable, pH-independent conjugates. Furthermore, we demonstrate that the chemically modified antibodies maintain antigen-binding capabilities. We apply this novel conjugation strategy to develop a surface-enhanced Raman spectroscopy (SERS)-based assay for the accurate subtyping of avian influenza viruses.


Asunto(s)
Anticuerpos/química , Técnicas Biosensibles , Oro , Nanopartículas del Metal , Animales , Pollos , Perros , Inmunoensayo , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N8 del Virus de la Influenza A , Virus de la Influenza A/clasificación , Células de Riñón Canino Madin Darby , Nanoconjugados
12.
Analyst ; 141(12): 3851-7, 2016 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-27113720

RESUMEN

The ability to evaluate antibody immobilization onto gold nanoparticles is critical for assessing coupling chemistry and optimizing the sensitivity of nanoparticle-enabled biosensors. Herein, we developed a fluorescence-based method for directly quantifying antibodies bound onto gold nanoparticles. Antibody-modified gold nanoparticles were treated with KI/I2 etchant to dissolve the gold nanoparticles. A desalting spin column was used to recover the antibody released from the nanoparticles, and NanoOrange, a fluorescent dye, was used to quantify the antibody. We determined 309 ± 93 antibodies adsorb onto a 60 nm gold nanoparticles (2.6 × 10(10) NP mL(-1)), which is consistent with a fully adsorbed monolayer based on the footprint of an IgG molecule. Moreover, the increase in hydrodynamic diameter of the conjugated nanoparticle (76 nm) compared to that of the unconjugated nanoparticle (62 nm) confirmed that multilayers did not form. A more conventional method of indirectly quantifying the adsorbed antibody by analysis of the supernatant overestimated the antibody surface coverage (660 ± 87 antibodies per nanoparticle); thus we propose the method described herein as a more accurate alternative to the conventional approach.

13.
Langmuir ; 30(22): 6309-13, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24854627

RESUMEN

Gold nanoparticle (AuNP)-Au film constructs were prepared using antibody-antigen interactions or a small organic cross-linker to systematically control the gap between the AuNP and Au film. Surface-enhanced Raman spectroscopy (SERS), scanning electron micrsocopy (SEM), and atomic force microscopy (AFM) were used to characterize each construct and elucidate structure-activity relationships. Interestingly, plasmonic coupling and SERS intensity were reversibly modulated with wetting/drying cycles for the protein immobilized AuNP, and this effect was attributed to changes in protein size with hydration state. This work provides insight into fundamental limitations of AuNP-enabled SERS bioassays and will facilitate rational design of novel biospecific ligands that maximize SERS sensitivity.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Nanopartículas del Metal/ultraestructura , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Espectrometría Raman
14.
ACS Sens ; 9(7): 3496-3501, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-38913420

RESUMEN

Here, we describe a SERS-based vertical flow assay as a platform technology suitable for point-of-care (POC) diagnostic testing. A capture substrate is constructed from filter paper embedded with spherical gold nanoparticles (AuNPs) and functionalized with an appropriate capture antibody. The capture substrate is loaded into a filtration device and connected to a syringe to rapidly and repeatedly pass the sample through the sensor for efficient antigen binding. The antigen is then labeled with a SERS-active detection probe. We show that only a few Raman reporter molecules, exclusively located adjacent to the plasmonic capture substrate, generate detectible signals. To maximize the signal from underutilized Raman reporter molecules, we employ a secondary signal enhancing probe that undergoes antibody-directed assembly to form plasmonic core-satellites. This facile enhancement step provides a 3.5-fold increase in the signal and a detection limit of 0.23 ng/mL (1.6 pM) for human IgG. This work highlights the potential to rationally design plasmonic architectures using widely available and reproducible spherical AuNPs to achieve large SERS enhancements for highly sensitive POC diagnostics.


Asunto(s)
Oro , Inmunoglobulina G , Nanopartículas del Metal , Espectrometría Raman , Espectrometría Raman/métodos , Oro/química , Nanopartículas del Metal/química , Inmunoensayo/métodos , Humanos , Inmunoglobulina G/inmunología , Límite de Detección , Anticuerpos/inmunología , Anticuerpos/química
15.
Anal Chem ; 85(18): 8609-17, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-23972208

RESUMEN

A rapid and simple SERS-based immunoassay has been developed to overcome diffusion-limited binding kinetics that often impedes rapid analysis in conventional heterogeneous immunoassays. This paper describes the development of an antibody-modified membrane as a flow-through capture substrate for a nanoparticle-enabled SERS immunoassay to enhance antibody-antigen binding kinetics. A thin layer of gold is plated onto polycarbonate track-etched nanoporous membranes via electroless deposition. Capture antibody is immobilized onto the surface of a gold-plated membrane via thiolate coupling chemistry to serve as a capture substrate. A syringe is then used to actively transport the analyte and extrinsic Raman labels to the capture substrate. The fabrication of the gold-plated membrane is thoroughly investigated and established as a viable capture substrate for a SERS-based immunoassay in the absence of sample/SERS label flow. A syringe pump is used to systematically investigate the effect of flow rate on antibody-antigen binding kinetics and demonstrate that active transport to the capture membrane surface expedites antibody-antigen binding. Mouse IgG and goat anti-mouse IgG are selected as a model antigen-antibody system to establish proof of principle. It is demonstrated that the assay for mouse IgG is reduced from 24 h to 10 min and a 10-fold improvement in detection limit is achieved with the flow assay developed herein relative to the passive, i.e., no flow, assay. Moreover, mouse serum is directly analyzed and IgG level is determined using the flow assay.


Asunto(s)
Oro/química , Inmunoglobulina G/sangre , Nanopartículas del Metal/química , Espectrometría Raman/métodos , Animales , Bovinos , Humanos , Inmunoensayo , Ratones
16.
Analyst ; 138(4): 1212-8, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23304695

RESUMEN

Protein-conjugated gold nanoparticles (AuNPs) have been extensively explored for the development of many novel protein assays. In this article, we demonstrate that nanoparticle tracking analysis (NTA) can be used as a rapid and simple analytical tool to monitor bioconjugation and to study protein-protein interactions. The adsorption of protein A onto gold nanoparticles was analyzed using NTA. The conjugation resulted in a measurable increase in hydrodynamic radius that correlated with protein A concentration, allowing conditions for complete conjugation to be elucidated. NTA was then used to investigate the binding of mouse IgG to protein A-conjugated AuNPs and the K(a) was measured as 2.00 × 10(7) M(-1). Furthermore, an assay for the detection of mouse IgG was developed using NTA to detect the binding to antibody-AuNP conjugates. This assay provided a detection limit of 3.2 ng mL(-1); however, the formation of aggregates resulting from the use of a polyclonal antibody and multiple binding sites on the antigen prevented the determination of binding affinity for this antibody-antigen system. To measure the binding affinity for this antibody-antigen system the IgG antigen was conjugated to the AuNPs and NTA was used to monitor the binding of the antibody. In this configuration aggregation of conjugates was not detected and a binding affinity constant of 2.80 × 10(8) M(-1) was measured. NTA results obtained in this work were validated by comparison to DLS. This work represents the first evaluation of NTA as an analytical tool for characterizing AuNP bioconjugates, investigating protein-protein binding, and detecting low levels of antigen in a bioassay.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Dispersión de Radiación , Animales , Bovinos , Oro/metabolismo , Inmunoglobulina G/análisis , Inmunoglobulina G/metabolismo , Ratones , Unión Proteica/fisiología , Albúmina Sérica Bovina/análisis , Albúmina Sérica Bovina/metabolismo
17.
J Am Chem Soc ; 134(31): 12889-92, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22788749

RESUMEN

Label-free surface-enhanced Raman spectroscopy (SERS) detection of nucleic acid hybridization is impeded by poor spectral reproducibility and the fact that the chemical signatures of hybridized and unhybridized sequences are highly similar. To overcome these issues, highly reproducible silver nanorod SERS substrates along with a straightforward least-squares (LS) technique have been employed for the quantitative determination of the relative ratios of the four nucleotide components A, C, G, and T/U before and after hybridization using a clinically relevant micro-RNA sequence.


Asunto(s)
MicroARNs/química , Hibridación de Ácido Nucleico , Espectrometría Raman/métodos , Análisis de los Mínimos Cuadrados , Nanotubos , Plata/química
18.
Appl Environ Microbiol ; 78(6): 1930-5, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22210215

RESUMEN

Mycoplasma gallisepticum is a bacterial pathogen of poultry that is estimated to cause annual losses exceeding $780 million. The National Poultry Improvement Plan guidelines recommend regular surveillance and intervention strategies to contain M. gallisepticum infections and ensure mycoplasma-free avian stocks, but several factors make detection of M. gallisepticum and diagnosis of M. gallisepticum infection a major challenge. Current techniques are laborious, require special expertise, and are typically plagued by false results. In this study, we describe a novel detection strategy which uses silver nanorod array-surface-enhanced Raman spectroscopy (NA-SERS) for direct detection of avian mycoplasmas. As a proof of concept for use in avian diagnostics, we used NA-SERS to detect and differentiate multiple strains of avian mycoplasma species, including Acholeplasma laidlawii, Mycoplasma gallinarum, Mycoplasma gallinaceum, Mycoplasma synoviae, and M. gallisepticum, including vaccine strains 6/85, F, and ts-11. Chemometric multivariate analysis of spectral data was used to classify these species rapidly and accurately, with >93% sensitivity and specificity. Furthermore, NA-SERS had a lower limit of detection that was 100-fold greater than that of standard PCR and comparable to that of real-time quantitative PCR. Detection of M. gallisepticum in choanal cleft swabs from experimentally infected birds yielded good sensitivity and specificity, suggesting that NA-SERS is applicable for clinical detection.


Asunto(s)
Acholeplasma/aislamiento & purificación , Técnicas Bacteriológicas/métodos , Aves/microbiología , Mycoplasma/aislamiento & purificación , Nanotubos , Plata/metabolismo , Espectrometría Raman/métodos , Acholeplasma/química , Acholeplasma/clasificación , Animales , Análisis por Micromatrices/métodos , Mycoplasma/química , Mycoplasma/clasificación , Sensibilidad y Especificidad
19.
Colloids Surf B Biointerfaces ; 220: 112867, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36182820

RESUMEN

A detailed understanding of protein-nanoparticle interactions is critical to realize the full potential of bioconjugate-enabled technologies. Parameters that lead to conformational changes in protein structure upon adsorption must be identified and controlled to mitigate loss of biological function. We hypothesized that the installation of thiol functional groups on a protein will facilitate robust adsorption to gold nanoparticles (AuNPs) and prevent protein unfolding to achieve thermodynamic stability. Here we investigated the adsorption behavior of α-chymotrypsin (ChT) and a thiolated analog of α-chymotrypsin (T-ChT) with AuNPs. ChT, which does not present any free thiols, was modified with 2-iminothiolane (Traut's reagent) to synthesize T-ChT consisting of two free thiols. Protein adsorption to AuNPs was monitored with dynamic light scattering and UV-vis spectrophotometry, and fluorescence spectra were acquired to assess changes in protein structure induced by interaction with the AuNP. The biological function of ChT, T-ChT, and respective bioconjugates were compared using a colorimetric enzymatic assay. The thiolated analog exhibited a greater affinity for the AuNP than the unmodified ChT, as determined from adsorption isotherms. The ChT protein formed a soft protein corona in which the enzyme denatures with prolonged exposure to AuNPs and, subsequently, lost enzymatic function. Conversely, the T-ChT formed a robust hard corona on the AuNP and retained structure and function. These data support the hypothesis, provide further insight into protein-AuNP interactions, and identify a simple chemical approach to synthesize robust and functional conjugates.


Asunto(s)
Oro , Nanopartículas del Metal , Oro/química , Nanopartículas del Metal/química , Quimotripsina/química , Adsorción , Compuestos de Sulfhidrilo , Proteínas
20.
Langmuir ; 27(19): 12253-60, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21877693

RESUMEN

In this article, a methacrylate-based spiropyran-containing copolymer was used as a colorimetric sensor to identify multiple metal ions simultaneously. Through UV-vis absorption spectroscopy, the relative binding affinity of merocyanine to each metal ion was investigated by displacement studies of a bound metal ion with a second metal ion of a higher binding affinity. We also show that because each metal ion gives rise to a distinct spectral response, partial least-squares discriminant analysis (PLS-DA) can be used to analyze the UV-vis absorbance spectra to identify the two metal ions that are present in solution at varying concentrations simply by dipping a coated polymer substrate into solution after irradiation. Partial least-squares regression analysis (PLS) was used to determine the metal ions in solution for several binary mixtures quantitatively. We also demonstrate that the quantitative determination depends on the relative binding preference of merocyanine to each metal ion.


Asunto(s)
Benzopiranos/química , Indoles/química , Membranas Artificiales , Metales Pesados/análisis , Nitrocompuestos/química , Iones/análisis , Análisis de los Mínimos Cuadrados , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA