Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Microbiol Spectr ; 12(4): e0358623, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38391232

RESUMEN

Although smallpox has been eradicated, other orthopoxviruses continue to be a public health concern as exemplified by the ongoing Mpox (formerly monkeypox) global outbreak. While medical countermeasures (MCMs) previously approved by the Food and Drug Administration for the treatment of smallpox have been adopted for Mpox, previously described vulnerabilities coupled with the questionable benefit of at least one of the therapeutics during the 2022 Mpox outbreak reinforce the need for identifying and developing other MCMs against orthopoxviruses. Here, we screened a panel of Merck proprietary small molecules and identified a novel nucleoside inhibitor with potent broad-spectrum antiviral activity against multiple orthopoxviruses. Efficacy testing of a 7-day dosing regimen of the orally administered nucleoside in a murine model of severe orthopoxvirus infection yielded a dose-dependent increase in survival. Treated animals had greatly reduced lesions in the lung and nasal cavity, particularly in the 10 µg/mL dosing group. Viral levels were also markedly lower in the UMM-766-treated animals. This work demonstrates that this nucleoside analog has anti-orthopoxvirus efficacy and can protect against severe disease in a murine orthopox model.IMPORTANCEThe recent monkeypox virus pandemic demonstrates that members of the orthopoxvirus, which also includes variola virus, which causes smallpox, remain a public health issue. While currently FDA-approved treatment options exist, risks that resistant strains of orthopoxviruses may arise are a great concern. Thus, continued exploration of anti-poxvirus treatments is warranted. Here, we developed a template for a high-throughput screening assay to identify anti-poxvirus small-molecule drugs. By screening available drug libraries, we identified a compound that inhibited orthopoxvirus replication in cell culture. We then showed that this drug can protect animals against severe disease. Our findings here support the use of existing drug libraries to identify orthopoxvirus-targeting drugs that may serve as human-safe products to thwart future outbreaks.


Asunto(s)
Mpox , Orthopoxvirus , Viruela , Virus de la Viruela , Animales , Ratones , Humanos , Nucleósidos/uso terapéutico , Viruela/tratamiento farmacológico , Viruela/prevención & control , Modelos Animales de Enfermedad
2.
Viruses ; 14(5)2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35632807

RESUMEN

A hallmark of severe acute respiratory syndrome virus (SARS-CoV-2) replication is the discontinuous transcription of open reading frames (ORFs) encoding structural virus proteins. Real-time reverse transcription PCR (RT-qPCR) assays in previous publications used either single or multiplex assays for SARS-CoV-2 genomic RNA detection and a singleplex approach for subgenomic RNA detection. Although multiplex approaches often target multiple genomic RNA segments, an assay that concurrently detects genomic and subgenomic targets has been lacking. To bridge this gap, we developed two duplex one-step RT-qPCR assays that detect SARS-CoV-2 genomic ORF1a and either subgenomic spike or subgenomic ORF3a RNAs. All primers and probes for our assays were designed to bind to variants of SARS-CoV-2. In this study, our assays successfully detected SARS-CoV-2 Washington strain and delta variant isolates at various time points during the course of live virus infection in vitro. The ability to quantify subgenomic SARS-CoV-2 RNA is important, as it may indicate the presence of active replication, particularly in samples collected longitudinally. Furthermore, specific detection of genomic and subgenomic RNAs simultaneously in a single reaction increases assay efficiency, potentially leading to expedited lucidity about viral replication and pathogenesis of any variant of SARS-CoV-2.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/diagnóstico , Genómica , Humanos , ARN Viral/análisis , ARN Viral/genética , SARS-CoV-2/genética
3.
Front Cell Infect Microbiol ; 12: 798978, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35463647

RESUMEN

Junín virus (JUNV), a New World arenavirus, is a rodent-borne virus and the causative agent of Argentine hemorrhagic fever. Humans become infected through exposure to rodent host secreta and excreta and the resulting infection can lead to an acute inflammatory disease with significant morbidity and mortality. Little is understood about the molecular pathogenesis of arenavirus hemorrhagic fever infections. We utilized Reverse Phase Protein Microarrays (RPPA) to compare global alterations in the host proteome following infection with an attenuated vaccine strain, Candid#1 (CD1), and the most parental virulent strain, XJ13, of JUNV in a human cell culture line. Human small airway epithelial cells were infected with CD1 or XJ13 at an MOI of 10, or mock infected. To determine proteomic changes at early timepoints (T = 1, 3, 8 and 24 h), the JUNV infected or mock infected cells were lysed in compatible buffers for RPPA. Out of 113 proteins that were examined by RPPA, 14 proteins were significantly altered following JUNV infection. Several proteins were commonly phosphorylated between the two strains and these correspond to entry and early replication events, to include p38 mitogen-activated protein kinase (MAPK), heat shock protein 27 (HSP27), and nuclear factor kappa B (NFκB). We qualitatively confirmed the alterations of these three proteins following infection by western blot analysis. We also determined that the inhibition of either p38 MAPK, with the small molecule inhibitor SB 203580 or siRNA knockdown, or HSP27, by siRNA knockdown, significantly decreases JUNV replication. Our data suggests that HSP27 phosphorylation at S82 upon virus infection is dependent on p38 MAPK activity. This work sheds light on the nuances of arenavirus replication.


Asunto(s)
Fiebre Hemorrágica Americana , Virus Junin , Proteínas de Choque Térmico HSP27 , Humanos , Virus Junin/genética , Proteómica , ARN Interferente Pequeño/genética , Proteínas Quinasas p38 Activadas por Mitógenos
4.
iScience ; 23(4): 100999, 2020 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-32252021

RESUMEN

The 2014-2016 West Africa Ebola virus (EBOV) outbreak coupled with the most recent outbreaks in Central Africa underscore the need to develop effective treatment strategies against EBOV. Although several therapeutic options have shown great potential, developing a wider breadth of countermeasures would increase our efforts to combat the highly lethal EBOV. Here we show that human cathelicidin antimicrobial peptide (AMP) LL-37 and engineered LL-37 AMPs inhibit the infection of recombinant virus pseudotyped with EBOV glycoprotein (GP) and the wild-type EBOV. These AMPs target EBOV infection at the endosomal cell-entry step by impairing cathepsin B-mediated processing of EBOV GP. Furthermore, two engineered AMPs containing D-amino acids are particularly potent in blocking EBOV infection in comparison with other AMPs, most likely owing to their resistance to intracellular enzymatic degradation. Our results identify AMPs as a novel class of anti-EBOV therapeutics and demonstrate the feasibility of engineering AMPs for improved therapeutic efficacy.

5.
Eur J Med Chem ; 162: 32-50, 2019 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-30408747

RESUMEN

Ebola virus (EBOV) causes a deadly hemorrhagic fever in humans and non-human primates. There is currently no FDA-approved vaccine or medication to counter this disease. Here, we report on the design, synthesis and anti-viral activities of two classes of compounds which show high potency against EBOV in both in vitro cell culture assays and in vivo mouse models Ebola viral disease. These compounds incorporate the structural features of cationic amphiphilic drugs (CAD), i.e they possess both a hydrophobic domain and a hydrophilic domain consisting of an ionizable amine functional group. These structural features enable easily diffusion into cells but once inside an acidic compartment their amine groups became protonated, ionized and remain trapped inside the acidic compartments such as late endosomes and lysosomes. These compounds, by virtue of their lysomotrophic functions, blocked EBOV entry. However, unlike other drugs containing a CAD moiety including chloroquine and amodiaquine, compounds reported in this study display faster kinetics of accumulation in the lysosomes, robust expansion of late endosome/lysosomes, relatively more potent suppression of lysosome fusion with other vesicular compartments and inhibition of cathepsins activities, all of which play a vital role in anti-EBOV activity. Furthermore, the diazachrysene 2 (ZSML08) that showed most potent activity against EBOV in in vitro cell culture assays also showed significant survival benefit with 100% protection in mouse models of Ebola virus disease, at a low dose of 10 mg/kg/day. Lastly, toxicity studies in vivo using zebrafish models suggest no developmental defects or toxicity associated with these compounds. Overall, these studies describe two new pharmacophores that by virtue of being potent lysosomotrophs, display potent anti-EBOV activities both in vitro and in vivo animal models of EBOV disease.


Asunto(s)
Antivirales/química , Crisenos/química , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Animales , Antivirales/farmacología , Antivirales/toxicidad , Crisenos/farmacología , Crisenos/toxicidad , Lisosomas/metabolismo , Ratones , Tensoactivos , Internalización del Virus/efectos de los fármacos , Pez Cebra
6.
PLoS Pathog ; 12(2): e1005437, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26837067

RESUMEN

Activated protein kinase R (PKR) plays a vital role in antiviral defense primarily by inhibiting protein synthesis and augmenting interferon responses. Many viral proteins have adopted unique strategies to counteract the deleterious effects of PKR. The NSs (Non-structural s) protein which is encoded by Rift Valley fever virus (RVFV) promotes early PKR proteasomal degradation through a previously undefined mechanism. In this study, we demonstrate that NSs carries out this activity by assembling the SCF (SKP1-CUL1-F-box)(FBXW11) E3 ligase. NSs binds to the F-box protein, FBXW11, via the six amino acid sequence DDGFVE called the degron sequence and recruits PKR through an alternate binding site to the SCF(FBXW11) E3 ligase. We further show that disrupting the assembly of the SCF(FBXW11-NSs) E3 ligase with MLN4924 (a small molecule inhibitor of SCF E3 ligase activity) or NSs degron viral mutants or siRNA knockdown of FBXW11 can block PKR degradation. Surprisingly, under these conditions when PKR degradation was blocked, NSs was essential and sufficient to activate PKR causing potent inhibition of RVFV infection by suppressing viral protein synthesis. These antiviral effects were antagonized by the loss of PKR expression or with a NSs deleted mutant virus. Therefore, early PKR activation by disassembly of SCF(FBXW11-NSs) E3 ligase is sufficient to inhibit RVFV infection. Furthermore, FBXW11 and BTRC are the two homologues of the ßTrCP (Beta-transducin repeat containing protein) gene that were previously described to be functionally redundant. However, in RVFV infection, among the two homologues of ßTrCP, FBXW11 plays a dominant role in PKR degradation and is the limiting factor in the assembly of the SCF(FBXW11) complex. Thus, FBXW11 serves as a master regulator of RVFV infection by promoting PKR degradation. Overall these findings provide new insights into NSs regulation of PKR activity and offer potential opportunities for therapeutic intervention of RVFV infection.


Asunto(s)
Proteínas F-Box/metabolismo , Virus de la Fiebre del Valle del Rift , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética , Animales , Antivirales/farmacología , Línea Celular , Proteínas Cullin/metabolismo , Genes Reguladores/genética , Humanos , Fosforilación/genética , Ubiquitina-Proteína Ligasas/metabolismo
7.
J Biomol Screen ; 20(1): 141-52, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25342145

RESUMEN

High-content image-based screening was developed as an approach to test a small-molecule library of compounds targeting signal transduction pathways for antiviral activity against multiple highly pathogenic RNA viruses. Of the 2843 compounds screened, 120 compounds exhibited ≥60% antiviral activity. Four compounds (E225-0969, E528-0039, G118-0778, and G544-0735), which were most active against Rift Valley fever virus (RVFV) and showed broad-spectrum antiviral activity, were selected for further evaluation for their concentration-response profile and cytotoxicity. These compounds did not show any visible cytotoxicity at the highest concentration of compound tested (200 µM). All four of these compounds were more active than ribavirin against several viruses. One compound, E225-0969, had the lowest effective concentration (EC50 = 1.9-8.92 µM) for all the viruses tested. This compound was 13- and 43-fold more inhibitory against RVFV and Chikungunya virus (CHIKV), respectively, than ribavirin. The highest selectivity index (>106.2) was for E225-0969 against CHIKV. Time-of-addition assays suggested that all four lead compounds targeted early steps in the viral life cycle (entry and/or replication) but not virus egress. Overall, this work demonstrates that high-content image analysis can be used to screen chemical libraries for new antivirals against highly pathogenic viruses.


Asunto(s)
Antivirales/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Pruebas de Sensibilidad Microbiana/métodos , Virus ARN/efectos de los fármacos , Virus ARN/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antivirales/química , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Ensayos Analíticos de Alto Rendimiento/normas , Humanos , Pruebas de Sensibilidad Microbiana/normas , Microscopía Fluorescente , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
8.
PLoS Negl Trop Dis ; 8(8): e3095, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25144302

RESUMEN

High content image-based screening was developed as an approach to test a protease inhibitor small molecule library for antiviral activity against Rift Valley fever virus (RVFV) and to determine their mechanism of action. RVFV is the causative agent of severe disease of humans and animals throughout Africa and the Arabian Peninsula. Of the 849 compounds screened, 34 compounds exhibited ≥ 50% inhibition against RVFV. All of the hit compounds could be classified into 4 distinct groups based on their unique chemical backbone. Some of the compounds also showed broad antiviral activity against several highly pathogenic RNA viruses including Ebola, Marburg, Venezuela equine encephalitis, and Lassa viruses. Four hit compounds (C795-0925, D011-2120, F694-1532 and G202-0362), which were most active against RVFV and showed broad-spectrum antiviral activity, were selected for further evaluation for their cytotoxicity, dose response profile, and mode of action using classical virological methods and high-content imaging analysis. Time-of-addition assays in RVFV infections suggested that D011-2120 and G202-0362 targeted virus egress, while C795-0925 and F694-1532 inhibited virus replication. We showed that D011-2120 exhibited its antiviral effects by blocking microtubule polymerization, thereby disrupting the Golgi complex and inhibiting viral trafficking to the plasma membrane during virus egress. While G202-0362 also affected virus egress, it appears to do so by a different mechanism, namely by blocking virus budding from the trans Golgi. F694-1532 inhibited viral replication, but also appeared to inhibit overall cellular gene expression. However, G202-0362 and C795-0925 did not alter any of the morphological features that we examined and thus may prove to be good candidates for antiviral drug development. Overall this work demonstrates that high-content image analysis can be used to screen chemical libraries for new antivirals and to determine their mechanism of action and any possible deleterious effects on host cellular biology.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Inhibidores de Proteasas/farmacología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Virología/métodos , Animales , Chlorocebus aethiops , Ebolavirus , Células HeLa , Humanos , Fiebre del Valle del Rift , Células Vero
9.
J Virol ; 87(15): 8451-64, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23720721

RESUMEN

We show that interferon-induced transmembrane protein 1 (IFITM-1), IFITM-2, and IFITM-3 exhibit a broad spectrum of antiviral activity against several members of the Bunyaviridae family, including Rift Valley fever virus (RVFV), La Crosse virus, Andes virus, and Hantaan virus, all of which can cause severe disease in humans and animals. We found that RVFV was restricted by IFITM-2 and -3 but not by IFITM-1, whereas the remaining viruses were equally restricted by all IFITMs. Indeed, at low doses of alpha interferon (IFN-α), IFITM-2 and -3 mediated more than half of the antiviral activity of IFN-α against RVFV. IFITM-2 and -3 restricted RVFV infection mostly by preventing virus membrane fusion with endosomes, while they had no effect on virion attachment to cells, endocytosis, or viral replication kinetics. We found that large fractions of IFITM-2 and IFITM-3 occupy vesicular compartments that are distinct from the vesicles coated by IFITM-1. In addition, although overexpression of all IFITMs expanded vesicular and acidified compartments within cells, there were marked phenotypic differences among the vesicular compartments occupied by IFITMs. Collectively, our data provide new insights into the possible mechanisms by which the IFITM family members restrict distinct viruses.


Asunto(s)
Antígenos de Diferenciación/inmunología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Virus de la Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Internalización del Virus , Animales , Línea Celular , Virus Hantaan/inmunología , Virus Hantaan/fisiología , Orthohantavirus/inmunología , Orthohantavirus/fisiología , Humanos , Interferón-alfa/inmunología , Virus La Crosse/inmunología , Virus La Crosse/fisiología
10.
Viruses ; 4(10): 1865-77, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-23202445

RESUMEN

Viruses modulate a number of host biological responses including the cell cycle to favor their replication. In this study, we developed a high-content imaging (HCI) assay to measure DNA content and identify different phases of the cell cycle. We then investigated the potential effects of cell cycle arrest on Ebola virus (EBOV) infection. Cells arrested in G1 phase by serum starvation or G1/S phase using aphidicolin or G2/M phase using nocodazole showed much reduced EBOV infection compared to the untreated control. Release of cells from serum starvation or aphidicolin block resulted in a time-dependent increase in the percentage of EBOV infected cells. The effect of EBOV infection on cell cycle progression was found to be cell-type dependent. Infection of asynchronous MCF-10A cells with EBOV resulted in a reduced number of cells in G2/M phase with concomitant increase of cells in G1 phase. However, these effects were not observed in HeLa or A549 cells. Together, our studies suggest that EBOV requires actively proliferating cells for efficient replication. Furthermore, multiplexing of HCI based assays to detect viral infection, cell cycle status and other phenotypic changes in a single cell population will provide useful information during screening campaigns using siRNA and small molecule therapeutics.


Asunto(s)
Afidicolina/farmacología , Puntos de Control del Ciclo Celular , Ciclo Celular/efectos de los fármacos , Ebolavirus/patogenicidad , Procesamiento de Imagen Asistido por Computador/métodos , Bencimidazoles/metabolismo , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/patología , Proliferación Celular , Medios de Cultivo/metabolismo , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/patología , Fiebre Hemorrágica Ebola/virología , Humanos , Nocodazol/farmacología , Suero/metabolismo , Factores de Tiempo , Replicación Viral
11.
Antiviral Res ; 93(1): 23-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22027648

RESUMEN

The highly lethal filoviruses, Ebola and Marburg cause severe hemorrhagic fever in humans and non-human primates. To date there are no licensed vaccines or therapeutics to counter these infections. Identifying novel pathways and host targets that play an essential role during infection will provide potential targets to develop therapeutics. Small molecule chemical screening for Ebola virus inhibitors resulted in identification of a compound NSC 62914. The compound was found to exhibit anti-filovirus activity in cell-based assays and in vivo protected mice following challenge with Ebola or Marburg viruses. Additionally, the compound was found to inhibit Rift Valley fever virus, Lassa virus and Venezuelan equine encephalitis virus in cell-based assays. Investigation of the mechanism of action of the compound revealed that it had antioxidant properties. Specifically, compound NSC 62914 was found to act as a scavenger of reactive oxygen species, and to up-regulate oxidative stress-induced genes. However, four known antioxidant compounds failed to inhibit filovirus infection, thus suggesting that the mechanistic basis of the antiviral function of the antioxidant NSC 62914 may involve modulation of multiple signaling pathways/targets.


Asunto(s)
Antivirales/farmacología , Compuestos de Bencidrilo/farmacología , Ebolavirus/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Marburgvirus/efectos de los fármacos , Fenoles/farmacología , Animales , Antivirales/química , Antivirales/uso terapéutico , Compuestos de Bencidrilo/química , Compuestos de Bencidrilo/toxicidad , Línea Celular Tumoral , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/química , Depuradores de Radicales Libres/uso terapéutico , Células HeLa , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento , Humanos , Enfermedad del Virus de Marburg/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Fenoles/química , Fenoles/toxicidad , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
12.
J Biomol Screen ; 15(7): 755-65, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20639507

RESUMEN

Filoviruses such as Ebola (EBOV) and Marburg (MARV) are single-stranded negative sense RNA viruses that cause acute hemorrhagic fever with high mortality rates. Currently, there are no licensed vaccines or therapeutics to counter filovirus infections in humans. The development of higher throughput/high-content primary screening assays followed by validation using the low-throughput traditional plaque or real-time PCR assays will greatly aid efforts toward the discovery of novel antiviral therapeutics. Specifically, high-content imaging technology is increasingly being applied for primary drug screening. In this study, the authors describe the challenges encountered when optimizing bioassays based on image acquisition and analyses for the highly pathogenic filoviruses Ebola and Marburg. A number of biological and imaging-related variables such as plating density, multiplicity of infection, the number of fields scanned per well, fluorescence intensity, and the cell number analyzed were evaluated during the development of these assays. Furthermore, the authors demonstrate the benefits related to the statistical analyses of single-cell data to account for heterogeneity in the subcellular localization and whole-cell integrated intensity of the viral antigen staining pattern. In conclusion, they show that image-based methods represent powerful screening tools for identifying antiviral compounds for highly pathogenic viruses.


Asunto(s)
Ebolavirus/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento/métodos , Imagenología Tridimensional/métodos , Marburgvirus/aislamiento & purificación , Animales , Antivirales/análisis , Antivirales/farmacología , Automatización , Chlorocebus aethiops , Ebolavirus/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Fiebre Hemorrágica Ebola/virología , Enfermedad del Virus de Marburg/virología , Marburgvirus/fisiología , Células Vero , Replicación Viral
13.
Anal Chem ; 81(10): 3896-902, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19374426

RESUMEN

A rapid and sensitive detection of staphylococcal enterotoxin B (SEB) was developed using a novel acoustic sensing technique: Resonant Acoustic Profiling (RAP), which utilizes high-frequency piezoelectric quartz resonators for monitoring biomolecular interactions. An automated four-channel instrument consisting of acoustic sensors covalently conjugated with anti-SEB antibodies was used. As the samples flowed across control and active sensors simultaneously, binding was measured as a change in the resonant frequency. The lower limit of detection (LLOD) for the label free direct format was 25 ng/mL. Detection sensitivity was increased by adding mass sequentially to the captured SEB on the sensor in the form of sandwich antibodies and biotin-avidin-based gold nanoparticles. The LLOD for the mass enhanced formats were 5 and 0.5 ng/mL of SEB, respectively. The lowest sensitivity corresponds to 1.3 fM in a 75 microL sample. The total assay time including the enhancement steps was less than 10 min. SEB was detected in both neat urine and PBS buffer-spiked samples, with linear correlations between resonant frequency signals and SEB concentrations (R(2) of 0.999 and 0.998, respectively). No significant cross-reactivity was observed with homologue toxins SEA, SED, and TSST, but some cross-reactivity was observed with the closely related toxin SEC(1) when we used a polyclonal antibody in the assay. SEC(1) cross-reactivity was not observed when a SEB-specific monoclonal antibody was employed in the assay. Thus the specificity of the assay presented here was dependent on the quality of the antibodies used. In addition to detection, we evaluated RAP's ability to measure the toxin in unknown samples rapidly by measuring the initial binding rate of the interaction, thereby further shortening the assay time to 6 min.


Asunto(s)
Técnicas Biosensibles/métodos , Enterotoxinas/análisis , Anticuerpos Antibacterianos/química , Anticuerpos Antibacterianos/inmunología , Anticuerpos Inmovilizados/química , Anticuerpos Inmovilizados/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Toxinas Bacterianas/análisis , Técnicas Biosensibles/instrumentación , Reacciones Cruzadas , Oro/química , Nanopartículas del Metal/química , Sensibilidad y Especificidad , Coloración y Etiquetado , Superantígenos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...