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










Base de dados
Intervalo de ano de publicação
1.
ACS Sens ; 9(3): 1401-1409, 2024 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-38380622

RESUMO

Here we present LUCOS (Luminescent Competition Sensor), a modular and broadly applicable bioluminescent diagnostic platform enabling the detection of both small molecules and protein biomarkers. The construction of LUCOS sensors entails the covalent and site-specific coupling of a bioluminescent sensor component to an analyte-specific antibody via protein G-mediated photoconjugation. Target detection is accomplished through intramolecular competition with a tethered analyte competitor for antibody binding. We established two variants of LUCOS: an inherent ratiometric LUCOSR variant and an intensiometric LUCOSI version, which can be used for ratiometric detection upon the addition of a split calibrator luciferase. To demonstrate the versatility of the LUCOS platform, sensors were developed for the detection of the small molecule cortisol and the protein biomarker NT-proBNP. Sensors for both targets displayed analyte-dependent changes in the emission ratio and enabled detection in the micromolar concentration range (KD,app = 16-92 µM). Furthermore, we showed that the response range of the LUCOS sensor can be adjusted by attenuating the affinity of the tethered NT-proBNP competitor, which enabled detection in the nanomolar concentration range (KD,app = 317 ± 26 nM). Overall, the LUCOS platform offers a highly versatile and easy method to convert commercially available monoclonal antibodies into bioluminescent biosensors that provide a homogeneous alternative for the competitive immunoassay.


Assuntos
Anticorpos Monoclonais , Luciferases , Imunoensaio/métodos
2.
Nat Commun ; 12(1): 4586, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321486

RESUMO

Heterogeneous immunoassays such as ELISA have become indispensable in modern bioanalysis, yet translation into point-of-care assays is hindered by their dependence on external calibration and multiple washing and incubation steps. Here, we introduce RAPPID (Ratiometric Plug-and-Play Immunodiagnostics), a mix-and-measure homogeneous immunoassay platform that combines highly specific antibody-based detection with a ratiometric bioluminescent readout. The concept entails analyte-induced complementation of split NanoLuc luciferase fragments, photoconjugated to an antibody sandwich pair via protein G adapters. Introduction of a calibrator luciferase provides a robust ratiometric signal that allows direct in-sample calibration and quantitative measurements in complex media such as blood plasma. We developed RAPPID sensors that allow low-picomolar detection of several protein biomarkers, anti-drug antibodies, therapeutic antibodies, and both SARS-CoV-2 spike protein and anti-SARS-CoV-2 antibodies. With its easy-to-implement standardized workflow, RAPPID provides an attractive, fast, and low-cost alternative to traditional immunoassays, in an academic setting, in clinical laboratories, and for point-of-care applications.


Assuntos
Anticorpos Antivirais/sangue , Teste Sorológico para COVID-19/métodos , COVID-19/diagnóstico , Imunoensaio/normas , Medições Luminescentes/normas , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/sangue , COVID-19/imunologia , COVID-19/virologia , Teste Sorológico para COVID-19/instrumentação , Calibragem , Proteínas de Ligação ao GTP/química , Genes Reporter , Humanos , Imunoconjugados/química , Limite de Detecção , Luciferases/genética , Luciferases/metabolismo , Testes Imediatos , SARS-CoV-2/genética
3.
Bioconjug Chem ; 31(3): 656-662, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-31909607

RESUMO

Bioluminescent antibodies represent attractive detection agents in both bioanalytical assays and imaging. Currently, their preparation relies on genetic fusion of luciferases to antibodies or nonspecific chemical conjugation strategies. Here, we report a generic method to generate well-defined covalent antibody-luciferase conjugates starting from commercially available monoclonal antibodies. Our approach uses fusion proteins consisting of the bright blue light-emitting luciferase NanoLuc (NL) and an Fc-binding protein domain (Gx) that can be photo-cross-linked to the antibody using UV light illumination. Green and red color variants were constructed by tight fusion of the NanoLuc with a green fluorescent acceptor domain and introduction of Cy3, respectively. To increase the already bright NanoLuc emission, tandem fusions were successfully developed in which the Gx domain is fused to two or three copies of the NanoLuc domain. The Gx-NL fusion proteins can be efficiently photo-cross-linked to all human immunoglobulin G (IgG) isotypes and most mammalian IgG's using 365 nm light, yielding antibodies with either one or two luciferase domains. The bioluminescent antibodies were successfully used in cell immunostaining and bioanalytical assays such as enzyme-linked immunosorbent assay (ELISA) and Western blotting.


Assuntos
Anticorpos Monoclonais/química , Proteínas de Bactérias/química , Luciferases/genética , Substâncias Luminescentes/química , Proteínas Recombinantes de Fusão/química , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Humanos , Proteínas Recombinantes de Fusão/genética
4.
ACS Sens ; 4(1): 20-25, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30525479

RESUMO

Bioluminescent sensor proteins provide attractive tools for applications ranging from in vivo imaging to point-of-care testing. Here we introduce a new class of ratiometric bioluminescent sensor proteins that do not rely on direct modulation of BRET efficiency, but are based on competitive intramolecular complementation of split NanoLuc luciferase. Proof of concept for the feasibility of this sensor principle was provided by developing a blue-red light emitting sensor protein for the detection of anti-HIV1-p17 antibodies with a 500% change in emission ratio and a Kd of 10 pM. The new sensor design also improved the dynamic response of a sensor for the therapeutic antibody cetuximab 4-fold, allowing the direct quantification of this anti-EGFR antibody in undiluted blood plasma. The modular sensor architecture allows easy and systematic tuning of a sensor's dynamic range and should be generally applicable to allow rational engineering of bioluminescent sensor proteins.


Assuntos
Anticorpos Anti-HIV/sangue , Luciferases/química , Proteínas Luminescentes/química , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Cetuximab/imunologia , Corantes Fluorescentes/química , Anticorpos Anti-HIV/imunologia , Proteínas Luminescentes/imunologia , Estudo de Prova de Conceito
5.
Angew Chem Int Ed Engl ; 57(47): 15369-15373, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30168634

RESUMO

This work reports on fully integrated "sample-in-signal-out" microfluidic paper-based analytical devices (µPADs) relying on bioluminescence resonance energy transfer (BRET) switches for analyte recognition and colorimetric signal generation. The devices use BRET-based antibody sensing proteins integrated into vertically assembled layers of functionalized paper, and their design enables sample volume-independent and fully reagent-free operation, including on-device blood plasma separation. User operation is limited to the application of a single drop (20-30 µL) of sample (serum, whole blood) and the acquisition of a photograph 20 min after sample introduction, with no requirement for precise pipetting, liquid handling, or analytical equipment except for a camera. Simultaneous detection of three different antibodies (anti-HIV1, anti-HA, and anti-DEN1) in whole blood was achieved. Given its simplicity, this type of device is ideally suited for user-friendly point-of-care testing in low-resource environments.


Assuntos
Anticorpos Antivirais/sangue , Medições Luminescentes/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Papel , Animais , Colorimetria/instrumentação , Desenho de Equipamento , Humanos , Sistemas Automatizados de Assistência Junto ao Leito , Suínos , Viroses/sangue
6.
Anal Chem ; 90(5): 3592-3599, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29443503

RESUMO

Monitoring the levels of therapeutic antibodies in individual patients would allow patient-specific dose optimization, with the potential for major therapeutic and financial benefits. Our group recently developed a new platform of bioluminescent sensor proteins (LUMABS; LUMinescent AntiBody Sensor) that allow antibody detection directly in blood plasma. In this study, we targeted four clinically important therapeutic antibodies, the Her2-receptor targeting trastuzumab, the anti-CD20 antibodies rituximab and obinutuzumab, and the EGFR-blocking cetuximab. A strong correlation was found between the affinity of the antibody binding peptide and sensor performance. LUMABS sensors with physiologically relevant affinities and decent sensor responses were obtained for trastuzumab and cetuximab using mimotope and meditope peptides, respectively, with affinities in the 10-7 M range. The lower affinity of the CD20-derived cyclic peptide employed in the anti-CD20 LUMABS sensor ( Kd = 10-5 M), translated in a LUMABS sensor with a strongly attenuated sensor response. The trastuzumab and cetuximab sensors were further characterized with respect to binding kinetics and their performance in undiluted blood plasma. For both antibodies, LUMABS-based detection directly in plasma compared well to the analytical performance of commercial ELISA kits. Besides identifying important design parameters for the development of new LUMABS sensors, this work demonstrates the potential of the LUMABS platform for point-of-care detection of therapeutic antibodies.


Assuntos
Anticorpos Monoclonais Humanizados/sangue , Antineoplásicos Imunológicos/sangue , Cetuximab/sangue , Monitoramento de Medicamentos/métodos , Proteínas Luminescentes/análise , Rituximab/sangue , Trastuzumab/sangue , Técnicas Biossensoriais/métodos , Humanos , Termodinâmica
7.
ACS Sens ; 2(11): 1730-1736, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29037030

RESUMO

Single-step immunoassays that can be performed directly in solution are ideally suited for point-of-care diagnostics. Our group recently developed a new platform of bioluminescent sensor proteins (LUMABS; LUMinescent AntiBody Sensor) that allow antibody detection in blood plasma. Thus far, LUMABS has been limited to the detection of antibodies recognizing natural peptide epitopes. Here, we report the development of semisynthetic LUMABS sensors that recognize nonpeptide epitopes. The non-natural amino acid para-azidophenylalanine was introduced at the position of the original antibody-recognition sites as a chemical handle to enable site-specific conjugation of synthetic epitope molecules coupled to a dibenzocylcooctyne moiety via strain-promoted click chemistry. The approach was successfully demonstrated by developing semisynthetic LUMABS sensors for antibodies targeting the small molecules dinitrophenol and creatinine (DNP-LUMABS and CR-LUMABS) with affinities of 5.8 pM and 1.3 nM, respectively. An important application of these semisynthetic LUMABS is the detection of small molecules using a competitive assay format, which is demonstrated here for the detection of creatinine. Using a preassembled complex of CR-LUMABS and an anti-creatinine antibody, the detection of high micromolar concentrations of creatinine was possible both in buffer and in 1:1 diluted blood plasma. The use of semisynthetic LUMABS sensors significantly expands the range of antibody targets and enables the application of LUMABS sensors for the ratiometric bioluminescent detection of small molecules using a competitive immunoassay format.


Assuntos
Anticorpos/imunologia , Creatinina/análise , Dinitrofenóis/análise , Imunoensaio/métodos , Proteínas Luminescentes/química , Alcinos/química , Azidas/química , Creatinina/imunologia , Dinitrofenóis/imunologia , Epitopos/química , Epitopos/imunologia , Fenilalanina/análogos & derivados , Fenilalanina/química , Soluções
8.
ACS Sens ; 2(6): 729-734, 2017 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-28670623

RESUMO

FRET-based caspase activity probes have become important tools to monitor apoptotic cell signaling. However, their dependence on external illumination is incompatible with light sensitive cells and hampers applications that suffer from autofluorescence and light scattering. Here we report the development of three caspase sensor proteins based on Bioluminescence Resonance Energy Transfer (BRET) that retain the advantages of genetically encoded, ratiometric optical probes but do not require external illumination. These sensors consist of the bright and stable luciferase NanoLuc and the fluorescent protein mNeonGreen, fused together via a linker containing a recognition site for caspase-3, -8, or -9. In vitro characterization showed that each caspase sensor displayed a robust 10-fold decrease in BRET ratio upon linker cleavage, with modest caspase specificity. Importantly, whereas scattering and background fluorescence precluded FRET-based detection of intracellular caspase activity in plate-reader assays, such measurements could be easily performed using our caspase BRET sensors in a high throughput format. The brightness of the BRET sensors also enabled long-term single-cell imaging, allowing BRET-based recording of cell heterogeneity in caspase activity in a heterogenic cell population.

9.
Methods Enzymol ; 589: 87-114, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28336075

RESUMO

FRET-sensors have become important tools for intracellular imaging, but their dependence on external illumination presents some limitations, such as photobleaching and phototoxicity, which limit measurements over extended periods of time. Fluorescence measurements also suffer from autofluorescence and light scattering, which hampers in vivo imaging and measurements in strongly absorbing and scattering media such as blood. In principle, these issues can be resolved by using sensors based on bioluminescence resonance energy transfer (BRET). The recent development of brighter and more stable luciferases and the concomitant improvement in luciferase substrates have substantially decreased the sensitivity gap between fluorescence and bioluminescence. As a result, the application of BRET-sensors is no longer restricted to measurements on cell populations, but they can also be used for imaging of single living cells, and BRET has started to emerge as an attractive sensor format for point-of-care diagnostics. The aim of this chapter is to first provide a brief overview of the basic design principles for BRET-sensors. Next, important design considerations will be discussed in more detail by describing the development of three different classes of BRET-sensors, both from our own work and that of others. These examples are all based on the NanoLuc luciferase, a bright and very stable blue light-emitting luciferase developed by Promega that has quickly risen to prominence in the bioluminescence field.


Assuntos
Técnicas Biossensoriais/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Medições Luminescentes/métodos , Proteínas Luminescentes/análise , Animais , Humanos , Proteínas Luminescentes/genética , Imagem Óptica/métodos , Testes Imediatos , Engenharia de Proteínas/métodos , Smartphone
10.
Anal Chem ; 88(8): 4525-32, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27018236

RESUMO

Antibody detection is of fundamental importance in many diagnostic and bioanalytical assays, yet current detection techniques tend to be laborious and/or expensive. We present a new sensor platform (LUMABS) based on bioluminescence resonance energy transfer (BRET) that allows detection of antibodies directly in solution using a smartphone as the sole piece of equipment. LUMABS are single-protein sensors that consist of the blue-light emitting luciferase NanoLuc connected via a semiflexible linker to the green fluorescent acceptor protein mNeonGreen, which are kept close together using helper domains. Binding of an antibody to epitope sequences flanking the linker disrupts the interaction between the helper domains, resulting in a large decrease in BRET efficiency. The resulting change in color of the emitted light from green-blue to blue can be detected directly in blood plasma, even at picomolar concentrations of antibody. Moreover, the modular architecture of LUMABS allows changing of target specificity by simple exchange of epitope sequences, as demonstrated here for antibodies against HIV1-p17, hemagglutinin (HA), and dengue virus type I. The combination of sensitive ratiometric bioluminescent detection and the intrinsic modularity of the LUMABS design provides an attractive generic platform for point-of-care antibody detection that avoids the complex liquid handling steps associated with conventional immunoassays.


Assuntos
Anticorpos/sangue , Proteínas Luminescentes/análise , Proteínas Luminescentes/química , Smartphone , Transferência Ressonante de Energia de Fluorescência , Humanos , Proteínas Luminescentes/isolamento & purificação
11.
Org Biomol Chem ; 11(44): 7642-9, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24091607

RESUMO

Detection of antibodies is essential for the diagnosis of many disease states, including infectious diseases, autoimmune diseases and allergies. Most current antibody detection assays involve multistep detection schemes in which molecular recognition and signal generation are separate processes. A well-known example is the enzyme-linked immunosorbent assay (ELISA), which combines high sensitivity and specificity with strong signal amplification. However, ELISA and other heterogeneous methods require multiple, time-consuming washing and incubation steps, which limits their applicability in point-of-care diagnostics and high-throughput applications. In recent years, several new antibody detection strategies have been developed in which antibody binding and signal generation are integrated within a single biomolecular reporter. These strategies aim to rival ELISA in terms of sensitivity and specificity, while decreasing the time and effort required to perform an assay. Here, we review recent developments in this field according to their mechanism of action and discuss their advantages and limitations.


Assuntos
Fosfatase Alcalina/química , Anticorpos/análise , beta-Galactosidase/química , beta-Lactamases/química , Regulação Alostérica , Ensaio de Imunoadsorção Enzimática , Polarização de Fluorescência , Limite de Detecção , Soluções
12.
ACS Chem Biol ; 8(10): 2133-9, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23962156

RESUMO

Elucidation of subcellular signaling networks by multiparameter imaging is hindered by a lack of sensitive FRET pairs spectrally compatible with the classic CFP/YFP pair. Here, we present a generic strategy to enhance the traditionally poor sensitivity of red FRET sensors by developing self-associating variants of mOrange and mCherry that allow sensors to switch between well-defined on- and off states. Requiring just a single mutation of the mFruit domain, this new FRET pair improved the dynamic range of protease sensors up to 10-fold and was essential to generate functional red variants of CFP-YFP-based Zn(2+) sensors. The large dynamic range afforded by the new red FRET pair allowed simultaneous use of differently colored Zn(2+) FRET sensors to image Zn(2+) over a broad concentration range in the same cellular compartment.


Assuntos
Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Células HeLa , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Bibliotecas de Moléculas Pequenas , Zinco/química , Proteína Vermelha Fluorescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...