Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Biosensors (Basel) ; 13(3)2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: covidwho-2253293

RESUMEN

Antibody measurements play a central role in the diagnosis of many autoimmune and infectious diseases. One antibody detection technology, Luciferase Immunoprecipitation Systems (LIPS), utilizes genetically encoded recombinant luciferase antigen fusion proteins in an immunoglobulin capture format to generate robust antibody measurement with high diagnostic sensitivity and specificity. The LIPS technology has been highly useful in detecting antibodies for research diagnostics and the discovery of new autoantigens. The methodology of the assay requires immunoglobulin binding reagents such as protein A/G beads and washing steps to process the immune complex before antibody levels are measured by light production with a luminometer. Recently, simplified mix and read immunoassays based on split components of the nanoluciferase enzyme in a complementation format have been developed for antibody measurements without requiring immunoglobulin-capturing beads or washing steps. The mix and read immunoassays utilize two or three nanoluciferase fragments which when reconstituted via antigen-specific antibody binding generate a functional enzyme. At present, these split luciferase tests have been developed mainly for detecting SARS-CoV-2 antibodies. Here, we describe the traditional LIPS technology and compare it to the new split luciferase methodologies focusing on their technical features, strengths, limitations, and future opportunities for diagnostic research, and clinical applications.


Asunto(s)
COVID-19 , Humanos , COVID-19/diagnóstico , SARS-CoV-2 , Luciferasas/metabolismo , Inmunoensayo , Anticuerpos Antivirales
2.
J Biol Chem ; 299(4): 103028, 2023 04.
Artículo en Inglés | MEDLINE | ID: covidwho-2242974

RESUMEN

The emergence of SARS-CoV-2, which is responsible for the COVID-19 pandemic, has highlighted the need for rapid characterization of viral mechanisms associated with cellular pathogenesis. Viral UTRs represent conserved genomic elements that contribute to such mechanisms. Structural details of most CoV UTRs are not available, however. Experimental approaches are needed to allow for the facile generation of high-quality viral RNA tertiary structural models, which can facilitate comparative mechanistic efforts. By integrating experimental and computational techniques, we herein report the efficient characterization of conserved RNA structures within the 5'UTR of the HCoV-OC43 genome, a lab-tractable model coronavirus. We provide evidence that the 5'UTR folds into a structure with well-defined stem-loops (SLs) as determined by chemical probing and direct detection of hydrogen bonds by NMR. We combine experimental base-pair restraints with global structural information from SAXS to generate a 3D model that reveals that SL1-4 adopts a topologically constrained structure wherein SLs 3 and 4 coaxially stack. Coaxial stacking is mediated by short linker nucleotides and allows SLs 1 to 2 to sample different cojoint orientations by pivoting about the SL3,4 helical axis. To evaluate the functional relevance of the SL3,4 coaxial helix, we engineered luciferase reporter constructs harboring the HCoV-OC43 5'UTR with mutations designed to abrogate coaxial stacking. Our results reveal that the SL3,4 helix intrinsically represses translation efficiency since the destabilizing mutations correlate with increased luciferase expression relative to wildtype without affecting reporter mRNA levels, thus highlighting how the 5'UTR structure contributes to the viral mechanism.


Asunto(s)
Regiones no Traducidas 5' , Coronavirus Humano OC43 , ARN Viral , Coronavirus Humano OC43/genética , Luciferasas/genética , Dispersión del Ángulo Pequeño , Difracción de Rayos X , ARN Viral/genética
3.
Sci Rep ; 13(1): 1753, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: covidwho-2221874

RESUMEN

Proteases play critical roles in various biological processes, including apoptosis and viral infection. Several protease biosensors have been developed; however, obtaining a reliable signal from a very low level of endogenous protease activity remains a challenge. In this study, we developed a highly sensitive protease biosensor, named FlipNanoLuc, based on the Oplophorus gracilirostris NanoLuc luciferase. The flipped ß-strand was restored by protease activation and cleavage, resulting in the reconstitution of luciferase and enzymatic activity. By making several modifications, such as introducing NanoBiT technology and CL1-PEST1 degradation tag, the FlipNanoLuc-based protease biosensor system achieved more than 500-fold luminescence increase in the corresponding protease-overexpressing cells. We demonstrated that the FlipNanoLuc-based caspase sensor can be utilized for the detection of staurosporine-induced apoptosis with sixfold increase in luminescence. Furthermore, we also demonstrated that the FlipNanoLuc-based coronavirus 3CL-protease sensor can be used to detect human coronavirus OC43 with tenfold increase in luminescence and severe acute respiratory syndrome-coronavirus-2 infections with 20-fold increase in luminescence by introducing the stem-loop 1 sequence to prevent the virus inducing global translational shutdown.


Asunto(s)
Apoptosis , Técnicas Biosensibles , COVID-19 , Péptido Hidrolasas , Humanos , Caspasas , COVID-19/diagnóstico , Luciferasas , SARS-CoV-2
4.
Anal Chim Acta ; 1238: 340633, 2023 Jan 15.
Artículo en Inglés | MEDLINE | ID: covidwho-2120488

RESUMEN

The Coronavirus disease 2019 (COVID-19) pandemic brings great challenges to the public health and social economics around the world. As the pandemic continues and the mass vaccination goes on, monitoring the antibodies is particularly important for the epidemiological survey and vaccine assessment. Here, we developed a luciferase immunoprecipitation assay combined with an automated platform to detect anti-Receptor Binding Domain (RBD) antibody, where protein A and protein G modified magnetic beads were used to capture antibodies in serum samples and SARS-CoV-2 RBD was fused with Gaussia luciferase to label the captured target antibodies. The whole detection procedure can be completed within 20 min. The developed assay has proven up to 32 times more sensitive than ELISA for the detection of RBD antibodies. Furthermore, the results of the antibody detection of sera from vaccination as well as convalescence displayed good performance. The automated platform may provide a powerful tool for the control of COVID-19 pandemic by vaccination and the research of SARS-CoV-2 seroconversion.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Pandemias , COVID-19/diagnóstico , Luciferasas , Anticuerpos Antivirales
5.
Sci Rep ; 12(1): 17966, 2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: covidwho-2087314

RESUMEN

The gold-standard method to evaluate a functional antiviral immune response is to titer neutralizing antibodies (NAbs) against a viral pathogen. This is historically performed using an in vitro assay of virus-mediated infection, which requires BSL-3 facilities. As these are insufficient in Latin American countries, including Mexico, scant information is obtained locally about viral pathogens NAb, using a functional assay. An alternative solution to using a BSL-3 assay with live virus is to use a BSL-2-safe assay with a non-replicative pseudovirus. Pseudoviral particles can be engineered to display a selected pathogen's entry protein on their surface, and to deliver a reporter gene into target cells upon transduction. Here we comprehensively describe the first development of a BSL-2 safe NAbs-measuring functional assay in Mexico, based on the production of pseudotyped lentiviral particles. As proof-of-concept, the assay is based on Nanoluc luciferase-mediated luminescence measurements from target cells transduced with SARS-CoV-2 Spike-pseudotyped lentiviral particles. We applied the optimized assay in a BSL-2 facility to measure NAbs in 65 serum samples, which evidenced the assay with 100% sensitivity, 86.6% specificity and 96% accuracy. Overall, this is the first report of a BSL-2 safe pseudovirus-based functional assay developed in Mexico to measure NAbs, and a cornerstone methodology necessary to measure NAbs with a functional assay in limited resources settings.


Asunto(s)
Anticuerpos Neutralizantes , COVID-19 , Humanos , SARS-CoV-2 , Pruebas de Neutralización/métodos , Glicoproteína de la Espiga del Coronavirus/metabolismo , Anticuerpos Antivirales , México , Luciferasas/genética , Antivirales
6.
ACS Appl Bio Mater ; 5(11): 5174-5180, 2022 Nov 21.
Artículo en Inglés | MEDLINE | ID: covidwho-2076971

RESUMEN

The COVID-19 pandemic has increased public health vigilance worldwide. The coronavirus (SARS-CoV-2) can spread via aerosols, and droplet-borne viruses remain viable on nonliving surfaces for long duration. Hence, effective antiviral coatings are highly useful in eliminating viral persistence on nonliving surfaces. Although innovative antiviral coatings have been designed, conventional procedures for antiviral assays are generally laborious, time-consuming, and have a high limit of detection. In the present study, we report a rapid and highly sensitive method for evaluating antiviral coatings by measuring the luciferase activity derived from recombinant Sendai virus (SeV). The physicochemical characteristics of SeV, which has a single-stranded RNA genome encapsulated within a lipid envelope, allow us to exploit it as an indicator of the physicochemical potential of coating materials against enveloped RNA viruses in general. We demonstrate that SeV-based assay systems allow for the rapid and quantitative evaluation of the surface coatings composed of iodine solubilized in polyvinyl acetate. Additionally, we have investigated the effect of mucins, the dominant protein component of saliva, on the antiviral activity of surface coatings. The presence of mucins in the SeV suspension considerably rescues luciferase activity at the viral-surface interface, presumably due to mucin-mediated viral protection. Our findings provide insights into a procedure capable of the rapid evaluation and optimization of surface coatings, and suggest an important role of the mucin in the valid evaluation of antiviral agents.


Asunto(s)
Antivirales , Virus Sendai , Antivirales/farmacología , Luciferasas , Mucinas , Virus Sendai/efectos de los fármacos
7.
Molecules ; 27(19)2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: covidwho-2066284

RESUMEN

Three unique 5,6-seco-hexahydrodibenzopyrans (seco-HHDBP) machaeridiols A-C, reported previously from Machaerium Pers., have displayed potent activities against methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus faecium, and E. faecalis (VRE). In order to enrich the pipeline of natural product-derived antimicrobial compounds, a series of novel machaeridiol-based analogs (1-17) were prepared by coupling stemofuran, pinosylvin, and resveratrol legends with monoterpene units R-(-)-α-phellandrene, (-)-p-mentha-2,8-diene-1-ol, and geraniol, and their inhibitory activities were profiled against MRSA ATCC 1708, VRE ATCC 700221, and cancer signaling pathways. Compounds 5 and 11 showed strong in vitro activities with MIC values of 2.5 µg/mL and 1.25 µg/mL against MRSA, respectively, and 2.50 µg/mL against VRE, while geranyl analog 14 was found to be moderately active (MIC 5 µg/mL). The reduction of the double bonds of the monoterpene unit of compound 5 resulted in 17, which had the same antibacterial potency (MIC 1.25 µg/mL and 2.50 µg/mL) as its parent, 5. Furthermore, a combination study between seco-HHDBP 17 and HHDBP machaeriol C displayed a synergistic effect with a fractional inhibitory concentrations (FIC) value of 0.5 against MRSA, showing a four-fold decrease in the MIC values of both 17 and machaeriol C, while no such effect was observed between vancomycin and 17. Compounds 11 and 17 were further tested in vivo against nosocomial MRSA at a single intranasal dose of 30 mg/kg in a murine model, and both compounds were not efficacious under these conditions. Finally, compounds 1-17 were profiled against a panel of luciferase genes that assessed the activity of complex cancer-related signaling pathways (i.e., transcription factors) using T98G glioblastoma multiforme cells. Among the compounds tested, the geranyl-substituted analog 14 exhibited strong inhibition against several signaling pathways, notably Smad, Myc, and Notch, with IC50 values of 2.17 µM, 1.86 µM, and 2.15 µM, respectively. In contrast, the anti-MRSA actives 5 and 17 were found to be inactive (IC50 > 20 µM) across the panel of these cancer-signaling pathways.


Asunto(s)
Antiinfecciosos , Productos Biológicos , Staphylococcus aureus Resistente a Meticilina , Neoplasias , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antiinfecciosos/farmacología , Productos Biológicos/farmacología , Luciferasas , Ratones , Pruebas de Sensibilidad Microbiana , Monoterpenos/farmacología , Resveratrol/farmacología , Transducción de Señal , Factores de Transcripción , Vancomicina/farmacología
8.
Virol J ; 19(1): 126, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: covidwho-2053923

RESUMEN

BACKGROUND: Viral antigen detection test is the most common method used to detect viruses in the field rapidly. However, due to the low sensitivity, it can only be used as an auxiliary diagnosis method for virus infection. Improving sensitivity is crucial for developing more accurate viral antigen tests. Nano luciferase (Nluc) is a sensitive reporter that has not been used in virus detection. RESULTS: In this study, we produced an intracellularly Nluc labeled detection antibody (Nluc-ch2C5) and evaluated its ability to improve the detection sensitivity of respiratory syndrome coronavirus 2 (SARS-CoV-2) antigens. Compared with the traditional horse-radish peroxidase (HRP) labeled antibody (HRP-ch2C5), Nluc-ch2C5 was 41 times more sensitive for inactivated SARS-CoV-2 virus by sandwich chemiluminescence ELISA. Then we applied Nluc-ch2C5 to establish an automatic magnet chemiluminescence immune assay (AMCA) for the SARS-CoV-2 viral spike protein, the limit of detection was 68 pfu/reaction. The clinical sensitivity and specificity reached 75% (24/32) and 100% (48/48) using 32 PCR-positive and 48 PCR-negative swab samples for clinical evaluation, which is more sensitive than the commercial ELSA kit and colloid gold strip kit. CONCLUSIONS: Here, monoclonal antibody ch2C5 served as a model antibody and the SARS-CoV-2 served as a model pathogen. The Nluc labeled detecting antibody (Nluc-ch2C5) significantly improved the detection sensitivity of SARS-CoV-2 antigen. This labeling principle applies to other viral infections, so this labeling and test format could be expected to play an important role in detecting other virus antigens.


Asunto(s)
COVID-19 , SARS-CoV-2 , Antígenos Virales/análisis , COVID-19/diagnóstico , Prueba de COVID-19 , Humanos , Luciferasas/genética , Sensibilidad y Especificidad
9.
Curr Protoc ; 2(10): e521, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: covidwho-2047527

RESUMEN

Antibody detection assays are essential for evaluating immunity of individuals against a given virus, and this has been particularly relevant during the COVID-19 pandemic. Current serology assays either require a laboratory setting and take >1 hr (i.e., enzyme-linked immunosorbent assay [ELISA]) or are rapid but only qualitative in nature and cannot accurately track antibody levels over time (i.e., lateral flow assay [LFA]). Therefore, there is a need for development of a rapid and simple but also quantitative assay that can evaluate antibody levels in patients accurately over time. We have developed an assay that uses a split nanoluciferase fused to the spike or nucleocapsid proteins of the SARS-CoV-2 virus to enable luminescent-based detection of spike- or nucleocapsid-binding antibodies in serum, plasma, and whole blood samples. The resulting approach is simple, rapid, and quantitative and is highly amenable to low-/medium-throughput scale using plate-based assays, high-throughput scale using robotics, and point-of-care applications. In this article, we describe how to perform the assay in a laboratory setting using a plate reader or liquid-handling robotics and in a point-of-care setting using a handheld, battery-powered luminometer. Together, these assays allow antibody detection to be easily performed in multiple settings by simplifying and reducing assay time in a laboratory or clinical environment and by allowing for antibody detection in point-of-care, nonlaboratory settings. © 2022 Wiley Periodicals LLC. Basic Protocol: SARS-CoV-2 antibody detection using the split-luciferase assay on a medium-throughput scale with a laboratory luminometer Alternate Protocol 1: High-throughput-based protocol for SARS-CoV-2 antibody detection using a robotic platform Alternate Protocol 2: Point-of-care-based protocol for SARS-CoV-2 antibody detection using a handheld luminometer Support Protocol: Determining positive/negative cutoffs for test samples and standardizing the assay between days.


Asunto(s)
Técnicas Biosensibles , COVID-19 , Anticuerpos Antivirales/análisis , COVID-19/diagnóstico , Técnicas de Laboratorio Clínico/métodos , Humanos , Luciferasas , Proteínas de la Nucleocápside , Pandemias , SARS-CoV-2 , Sensibilidad y Especificidad
10.
Anal Chem ; 94(40): 13700-13709, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: covidwho-2036735

RESUMEN

Identification of protein-protein interactions (PPIs) that occur in various cellular processes helps to reveal their potential molecular mechanisms, and there is still an urgent need to develop the assays to explore PPIs in living subjects. Here, we reported a near-infrared split luciferase complementation assay (SLCA) with enhanced bioluminescence produced by cleaving a luciferase, Akaluc, for exploring and visualizing PPIs in living cells and live mice. Compared with the previously developed and widely used red SLCA based on split firefly luciferase (Fluc-SLCA), the signal intensities for PPI recognition in living cells and live mice of the Akaluc-SLCA increased by ∼3.79-fold and ∼18.06-fold in the measured condition, respectively. Additionally, the interactions between the nucleocapsid protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and cellular RNA processing proteins were identified, and the drug evaluation assays were also performed in living cells using Akaluc-SLCA. This study provides a new tool in the near-infrared region for the identification of PPIs in living cells and in vivo and new information for the understanding and treatment of SARS-CoV-2.


Asunto(s)
COVID-19 , Luciferasas de Luciérnaga , Animales , Evaluación de Medicamentos , Luciferasas/genética , Luciferasas de Luciérnaga/metabolismo , Ratones , Proteínas de la Nucleocápside , SARS-CoV-2
11.
J Vis Exp ; (186)2022 08 17.
Artículo en Inglés | MEDLINE | ID: covidwho-2024393

RESUMEN

In vitro transcribed messenger RNA (mRNA) vaccines have displayed enormous potential in fighting against the coronavirus disease 2019 (COVID-19) pandemic. Efficient and safe delivery systems must be included in the mRNA vaccines due to the fragile properties of mRNA. A self-assembled peptide-poloxamine nanoparticle (PP-sNp) gene delivery system is specifically designed for the pulmonary delivery of nucleic acids and displays promising capabilities in mediating successful mRNA transfection. Here, an improved method for preparing PP-sNp is described to elaborate on how the PP-sNp encapsulates Metridia luciferase (MetLuc) mRNA and successfully transfects cultured cells. MetLuc-mRNA is obtained by an in vitro transcription process from a linear DNA template. A PP-sNp is produced by mixing synthetic peptide/poloxamine with mRNA solution using a microfluidic mixer, allowing for the self-assembly of PP-sNp. The charge of PP-sNp is subsequently evaluated by measuring the zeta potential. Meanwhile, the polydispersity and hydrodynamic size of PP-sNp nanoparticles are measured using dynamic light scattering. The mRNA/PP-sNp nanoparticles are transfected into cultured cells, and supernatants from the cell culture are assayed for luciferase activity. The representative results demonstrate their capacity for in vitro transfection. This protocol may shed light on developing next-generation mRNA vaccine delivery systems.


Asunto(s)
COVID-19 , Nanopartículas , Células Cultivadas , Humanos , Luciferasas/genética , Péptidos/genética , ARN Mensajero/genética , Transfección , Vacunas Sintéticas , Vacunas de ARNm
12.
Viruses ; 14(8)2022 08 04.
Artículo en Inglés | MEDLINE | ID: covidwho-2024282

RESUMEN

The rapid transmission of measles poses a great challenge for measles elimination. Thus, rapid testing is required to screen the health status in the population during measles outbreaks. A pseudotype-based virus neutralisation assay was used to measure neutralising antibody titres in serum samples collected from healthcare workers in Sheffield during the measles outbreak in 2016. Vesicular stomatitis virus (VSV) pseudotypes bearing the haemagglutinin and fusion glycoproteins of measles virus (MeV) and carrying a luciferase marker gene were prepared; the neutralising antibody titre was defined as the dilution resulting in 90% reduction in luciferase activity. Spearman's correlation coefficients between IgG titres and neutralising antibody levels ranged from 0.40 to 0.55 (p < 0.05) or from 0.71 to 0.79 (p < 0.0001) when the IgG titres were obtained using different testing kits. In addition, the currently used vaccine was observed to cross-neutralise most circulating MeV genotypes. However, the percentage of individuals being "well-protected" was lower than 95%, the target rate of vaccination coverage to eliminate measles. These results demonstrate that the level of clinical protection against measles in individuals could be inferred by IgG titre, as long as a precise correlation has been established between IgG testing and neutralisation assay; moreover, maintaining a high vaccination coverage rate is still necessary for measles elimination.


Asunto(s)
Anticuerpos Neutralizantes , Sarampión , Anticuerpos Antivirales , Brotes de Enfermedades/prevención & control , Personal de Salud , Humanos , Inmunoglobulina G , Luciferasas , Sarampión/epidemiología , Sarampión/prevención & control , Vacuna Antisarampión , Vacunación
13.
Methods Enzymol ; 675: 351-381, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-2007357

RESUMEN

After more than two years, COVID-19 still represents a global health burden of unprecedented extent and assessing the degree of immunity of individuals against SARS-CoV-2 remains a challenge. Virus neutralization assays represent the gold standard for assessing antibody-mediated protection against SARS-CoV-2 in sera from recovered and/or vaccinated individuals. Neutralizing antibodies block the interaction of viral spike protein with human angiotensin-converting enzyme 2 (ACE2) receptor in vitro and prevent viral entry into host cells. Classical viral neutralization assays using full replication-competent viruses are restricted to specific biosafety level 3-certified laboratories, limiting their utility for routine and large-scale applications. We developed therefore a cell-fusion-based assay building on the interaction between viral spike and ACE2 receptor expressed on two different cell lines, substantially reducing biosafety risks associated with classical viral neutralization assays. This chapter describes this simple, sensitive, safe and cost-effective approach for rapid and high-throughput evaluation of SARS-CoV-2 neutralizing antibodies relying on high-affinity NanoLuc® luciferase complementation technology (HiBiT). When applied to a variety of standards and patient samples, this method yields highly reproducible results in 96-well, as well as in 384-well format. The use of novel NanoLuc® substrates with increased signal stability like Nano-Glo® Endurazine™ furthermore allows for high flexibility in assay set-up and full automatization of all reading processes. Lastly, the assay is suitable to evaluate the neutralizing capacity of sera against the existing spike variants, and potentially variants that will emerge in the future.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Fusión Celular , Humanos , Luciferasas , Pruebas de Neutralización/métodos , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo
14.
Microbiol Spectr ; 10(5): e0237922, 2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: covidwho-1992955

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the highly contagious agent responsible for the coronavirus disease 2019 (COVID-19) pandemic. An essential requirement for understanding SARS-CoV-2 biology and the impact of antiviral therapeutics is a robust method to detect the presence of the virus in infected cells or animal models. Despite the development and successful generation of recombinant (r)SARS-CoV-2-expressing fluorescent or luciferase reporter genes, knowledge acquired from their use in in vitro assays and/or in live animals is limited to the properties of the fluorescent or luciferase reporter genes. Herein, for the first time, we engineered a replication-competent rSARS-CoV-2 that expresses both fluorescent (mCherry) and luciferase (Nluc) reporter genes (rSARS-CoV-2/mCherry-Nluc) to overcome limitations associated with the use of a single reporter gene. In cultured cells, rSARS-CoV-2/mCherry-Nluc displayed similar viral fitness as rSARS-CoV-2 expressing single reporter fluorescent and luciferase genes (rSARS-CoV-2/mCherry and rSARS-CoV-2/Nluc, respectively) or wild-type (WT) rSARS-CoV-2, while maintaining comparable expression levels of both reporter genes. In vivo, rSARS-CoV-2/mCherry-Nluc has similar pathogenicity in K18 human angiotensin-converting enzyme 2 (hACE2) transgenic mice than rSARS-CoV-2 expressing individual reporter genes or WT rSARS-CoV-2. Importantly, rSARS-CoV-2/mCherry-Nluc facilitates the assessment of viral infection and transmission in golden Syrian hamsters using in vivo imaging systems (IVIS). Altogether, this study demonstrates the feasibility of using this novel bioreporter-expressing rSARS-CoV-2 for the study of SARS-CoV-2 in vitro and in vivo. IMPORTANCE Despite the availability of vaccines and antivirals, the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to ravage health care institutions worldwide. Previously, we generated replication-competent recombinant (r)SARS-CoV-2 expressing fluorescent or luciferase reporter proteins to track viral infection in vitro and/or in vivo. However, these rSARS-CoV-2 are restricted to express only a single fluorescent or a luciferase reporter gene, limiting or preventing their use in specific in vitro assays and/or in vivo studies. To overcome this limitation, we have engineered a rSARS-CoV-2 expressing both fluorescent (mCherry) and luciferase (Nluc) genes and demonstrated its feasibility to study the biology of SARS-CoV-2 in vitro and/or in vivo, including the identification and characterization of neutralizing antibodies and/or antivirals. Using rodent models, we visualized SARS-CoV-2 infection and transmission through in vivo imaging systems (IVIS).


Asunto(s)
COVID-19 , Cricetinae , Ratones , Animales , Humanos , Enzima Convertidora de Angiotensina 2/genética , SARS-CoV-2/genética , Replicación Viral , Antivirales/farmacología , Luciferasas/genética , Luciferasas/farmacología , Anticuerpos Neutralizantes , Ratones Transgénicos
15.
Nat Commun ; 13(1): 3716, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1984382

RESUMEN

The COVID-19 pandemic triggered the development of numerous diagnostic tools to monitor infection and to determine immune response. Although assays to measure binding antibodies against SARS-CoV-2 are widely available, more specific tests measuring neutralization activities of antibodies are immediately needed to quantify the extent and duration of protection that results from infection or vaccination. We previously developed a 'Serological Assay based on a Tri-part split-NanoLuc® (SATiN)' to detect antibodies that bind to the spike (S) protein of SARS-CoV-2. Here, we expand on our previous work and describe a reconfigured version of the SATiN assay, called Neutralization SATiN (Neu-SATiN), which measures neutralization activity of antibodies directly from convalescent or vaccinated sera. The results obtained with our assay and other neutralization assays are comparable but with significantly shorter preparation and run time for Neu-SATiN. As the assay is modular, we further demonstrate that Neu-SATiN enables rapid assessment of the effectiveness of vaccines and level of protection against existing SARS-CoV-2 variants of concern and can therefore be readily adapted for emerging variants.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Antivirales , Humanos , Luciferasas , Glicoproteínas de Membrana/metabolismo , Pruebas de Neutralización , Pandemias , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral
16.
J Med Virol ; 94(12): 6073-6077, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-1976740

RESUMEN

SARS-CoV-2 infection causes syncytial pneumocyte in patients and this has been considered as a defining feature of severe COVID-19 cases. Traditional methods of syncytia quantification require the morphology characterization of fused cells either with light microscope or fluorescent microscope, which is time-consuming and not accurate. Here we developed a rapid and sensitive coculture system measuring spike-induced syncytia by using NanoLuc complementation system. We found the formation of syncytia occurred rapidly after ACE2-expressing cells exposure to spike protein. In addition, we found furin cleavage as well as the cell surface protease TMPRSS2 enhanced syncytia formation. Finally, we showed that this coculture system can be used to test the ability of different compound to inhibit syncytia formation, thus providing a useful tool to screen anti-syncytial drugs.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Enzima Convertidora de Angiotensina 2 , Furina/metabolismo , Humanos , Luciferasas , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
17.
Genes Cells ; 27(8): 537-543, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: covidwho-1879030

RESUMEN

The ongoing COVID-19 pandemic is caused by SARS-CoV-2. Although several effective vaccines that target the Spike protein on the viral surface have been deployed, additional therapeutic agents are urgently needed. Here, we developed a system to measure the Spike protein function by quantifying cellular membrane fusion induced by the Spike protein. The system enables the evaluation of the effects of drugs and neutralizing antibodies against SARS-CoV-2 without using live viruses. Furthermore, the system characterizes membrane fusion activity of the Spike protein of each variant to reveal that Delta variant has more potent than Wuhan and Omicron. Our system could lead to develop high-throughput screening for drug candidates and neutralization antibodies that target virus entry and characterize Spike proteins from variants.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Humanos , Luciferasas , Fusión de Membrana , Pandemias , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo
18.
Biosens Bioelectron ; 209: 114226, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1767929

RESUMEN

Protein sensors based on allosteric enzymes responding to target binding with rapid changes in enzymatic activity are potential tools for homogeneous assays. However, a high signal-to-noise ratio (S/N) is difficult to achieve in their construction. A high S/N is critical to discriminate signals from the background, a phenomenon that might largely vary among serum samples from different individuals. Herein, based on the modularized luciferase NanoLuc, we designed a novel biosensor called NanoSwitch. This sensor allows direct detection of antibodies in 1 µl serum in 45 min without washing steps. In the detection of Flag and HA antibodies, NanoSwitches respond to antibodies with S/N ratios of 33-fold and 42-fold, respectively. Further, we constructed a NanoSwitch for detecting SARS-CoV-2-specific antibodies, which showed over 200-fold S/N in serum samples. High S/N was achieved by a new working model, combining the turn-off of the sensor with human serum albumin and turn-on with a specific antibody. Also, we constructed NanoSwitches for detecting antibodies against the core protein of hepatitis C virus (HCV) and gp41 of the human immunodeficiency virus (HIV). Interestingly, these sensors demonstrated a high S/N and good performance in the assays of clinical samples; this was partly attributed to the combination of off-and-on models. In summary, we provide a novel type of protein sensor and a working model that potentially guides new sensor design with better performance.


Asunto(s)
Técnicas Biosensibles , COVID-19 , Anticuerpos Antivirales , COVID-19/diagnóstico , Humanos , Luciferasas , SARS-CoV-2
19.
Proc Natl Acad Sci U S A ; 119(15): e2120913119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1758464

RESUMEN

SignificanceThe coronavirus main protease (Mpro) is required for viral replication. Here, we obtained the extended conformation of the native monomer of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Mpro by trapping it with nanobodies and found that the catalytic domain and the helix domain dissociate, revealing allosteric targets. Another monomeric state is termed compact conformation and is similar to one protomer of the dimeric form. We designed a Nanoluc Binary Techonology (NanoBiT)-based high-throughput allosteric inhibitor assay based on structural conformational change. Our results provide insight into the maturation, dimerization, and catalysis of the coronavirus Mpro and pave a way to develop an anticoronaviral drug through targeting the maturation process to inhibit the autocleavage of Mpro.


Asunto(s)
Antivirales , COVID-19 , Proteasas 3C de Coronavirus , Inhibidores de Proteasas , SARS-CoV-2 , Regulación Alostérica/efectos de los fármacos , Antivirales/química , Antivirales/farmacología , COVID-19/enzimología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Humanos , Luciferasas , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Conformación Proteica , Multimerización de Proteína
20.
Commun Biol ; 5(1): 212, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1735294

RESUMEN

Internalization of membrane proteins plays a key role in many physiological functions; however, highly sensitive and versatile technologies are lacking to study such processes in real-time living systems. Here we describe an assay based on bioluminescence able to quantify membrane receptor trafficking for a wide variety of internalization mechanisms such as GPCR internalization/recycling, antibody-mediated internalization, and SARS-CoV2 viral infection. This study represents an alternative drug discovery tool to accelerate the drug development for a wide range of physiological processes, such as cancer, neurological, cardiopulmonary, metabolic, and infectious diseases including COVID-19.


Asunto(s)
Descubrimiento de Drogas/métodos , Proteínas de la Membrana , Transporte de Proteínas/fisiología , Espectrometría de Fluorescencia/métodos , COVID-19 , Desarrollo de Medicamentos/métodos , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Nanotecnología , Receptores Acoplados a Proteínas G , SARS-CoV-2/química , SARS-CoV-2/metabolismo , Internalización del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA