Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biomed Sci ; 30(1): 41, 2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37316861

RESUMEN

BACKGROUND: Flavivirus causes many serious public health problems worldwide. However, licensed DENV vaccine has restrictions on its use, and there is currently no approved ZIKV vaccine. Development of a potent and safe flavivirus vaccine is urgently needed. As a previous study revealed the epitope, RCPTQGE, located on the bc loop in the E protein domain II of DENV, in this study, we rationally designed and synthesized a series of peptides based on the sequence of JEV epitope RCPTTGE and DENV/ZIKV epitope RCPTQGE. METHODS: Immune sera were generated by immunization with the peptides which were synthesized by using five copies of RCPTTGE or RCPTQGE and named as JEV-NTE and DV/ZV-NTE. Immunogenicity and neutralizing abilities of JEV-NTE or DV/ZV-NTE-immune sera against flavivirus were evaluated by ELISA and neutralization tests, respectively. Protective efficacy in vivo were determined by passive transfer the immune sera into JEV-infected ICR or DENV- and ZIKV-challenged AG129 mice. In vitro and in vivo ADE assays were used to examine whether JEV-NTE or DV/ZV-NTE-immune sera would induce ADE. RESULTS: Passive immunization with JEV-NTE-immunized sera or DV/ZV-NTE-immunized sera could increase the survival rate or prolong the survival time in JEV-challenged ICR mice and reduce the viremia levels significantly in DENV- or ZIKV-infected AG129 mice. Furthermore, neither JEV -NTE- nor DV/ZV-NTE-immune sera induced antibody-dependent enhancement (ADE) as compared with the control mAb 4G2 both in vitro and in vivo. CONCLUSIONS: We showed for the first time that novel bc loop epitope RCPTQGE located on the amino acids 73 to 79 of DENV/ZIKV E protein could elicit cross-neutralizing antibodies and reduced the viremia level in DENV- and ZIKV-challenged AG129 mice. Our results highlighted that the bc loop epitope could be a promising target for flavivirus vaccine development.


Asunto(s)
Infección por el Virus Zika , Virus Zika , Animales , Ratones , Ratones Endogámicos ICR , Anticuerpos Neutralizantes , Viremia , Sueros Inmunes , Epítopos , Factores de Transcripción
2.
Microbiol Spectr ; : e0485422, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36856407

RESUMEN

Chikungunya fever is a mosquito-transmitted infectious disease that induces rash, myalgia, and persistent incapacitating arthralgia. At present, no vaccines or antiviral therapies specific to Chikungunya virus (CHIKV) infection have been approved, and research is currently restricted to biosafety level 3 containment. CHIKV-like replicon particles (VRPs) are single-cycle infectious particles containing viral structure proteins, as well as a defective genome to provide a safe surrogate for living CHIKV to facilitate the testing of vaccines and antivirals. However, inefficient RNA transfection and the potential emergence of the competent virus through recombination in mammalian cells limit VRP usability. This study describes a transfection-free system for the safe packaging of CHIK VRP with all necessary components via transduction of mosquito cell lines using a single baculovirus vector. We observed the release of substantial quantities of mosquito cell-derived CHIK VRP (mos-CHIK VRP) from baculovirus-transduced mosquito cell lines. The VRPs were shown to recapitulate viral replication and subgenomic dual reporter expression (enhanced green fluorescent protein [eGFP] and luciferase) in infected host cells. Interestingly, the rapid expression kinetics of the VRP-expressing luciferase reporter (6 h) makes it possible to use mos-CHIK VRPs for the rapid quantification of VRP infection. Treatment with antivirals (suramin or 6-azauridine) or neutralizing antibodies (monoclonal antibodies [MAbs] or patient sera) was shown to inhibit mos-CHIK VRP infection in a dose-dependent manner. Ease of manufacture, safety, scalability, and high throughput make mos-CHIK VRPs a highly valuable vehicle for the study of CHIKV biology, the detection of neutralizing (NT) antibody activity, and the screening of antivirals against CHIKV. IMPORTANCE This study proposes a transfection-free system that enables the safe packaging of CHIK VRPs with all necessary components via baculovirus transduction. Those mosquito cell-derived CHIK VRP (mos-CHIK VRPs) were shown to recapitulate viral replication and subgenomic dual reporter (enhanced green fluorescent protein [eGFP] and luciferase) expression in infected host cells. Rapid expression kinetics of the VRP-expressing luciferase reporter (within hours) opens the door to using mos-CHIK VRPs for the rapid quantification of neutralizing antibody and antiviral activity against CHIKV. To the best of our knowledge, this is the first study to report a mosquito cell-derived alphavirus VRP system. Note that this system could also be applied to other arboviruses to model the earliest event in arboviral infection in vertebrates.

3.
Appl Microbiol Biotechnol ; 107(1): 219-232, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36434113

RESUMEN

The spread of chikungunya virus (CHIKV) is reaching pandemic levels, and vaccines and antivirals to control CHIKV infection have yet to be approved. Virus-like particles (VLPs), a self-assembled native multi-subunit protein structure, could potentially be used as an antigen for serological detection and vaccine development. In the current study, we describe the production of novel CHIKV VLPs from mosquitoes using a Baculovirus/Mosquito (BacMos) system in a simple Biosafety Level-2 laboratory. Substantial envelope and capsid protein secretions were detected in culture medium. Co-fractionation of CHIKV E2, E1, and capsid proteins via sucrose gradient ultracentrifugation provided evidence of VLP formation. Transmission electron microscopy and dynamic light scattering analysis revealed the formation of VLPs in the form of spherical particles with a diameter of roughly 40 nm in transduced cells and culture medium. VLP-based IgM capture ELISA in CHIKV patient sera revealed native epitopes on the VLPs. These non-purified VLPs were shown to act as an antigen in CHIKV-specific IgM capture ELISA. The immunization of CHIKV-VLPs alone in mice induced a balance CHIKV-specific IgG2a/IgG1 antibodies and neutralized antibody responses. The study provides support for the hypothesis that mosquito cell-derived CHIKV VLPs could serve as a novel antigen for serological detection and the development of vaccines against CHIKV infection. KEY POINTS: • CHIKV VLPs secreted from BacMos-CHIKV 26S-transduced mosquito cell. • This CHIKV VLPs potentially serve as an alternative capture antigen for MAC-ELISA. • Unadjuvanted CHIK VLPs induce CHIKV-specific IgG and NT responses in mice.


Asunto(s)
Fiebre Chikungunya , Virus Chikungunya , Culicidae , Ratones , Animales , Fiebre Chikungunya/prevención & control , Anticuerpos Antivirales , Inmunoglobulina M , Inmunoglobulina G , Proteínas de la Cápside
4.
Front Immunol ; 13: 852452, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309328

RESUMEN

Dengue is among the most rapidly spreading arboviral disease in the world. A low-cost, easy to use point-of-care diagnostic tool for the detection and differentiation of dengue virus serotypes could improve clinical management, disease prevention, epidemiological surveillance, and outbreak monitoring, particularly in regions where multiple serotypes co-circulate. Despite widespread deployment, no commercial dengue antigen diagnostic test has proven effective in differentiating among dengue virus serotypes. In the current study, we first established mAb pairs and developed a multiplex lateral flow immunoassay for the simultaneous detection of the dengue viral NS1 antigen and identification of serotype. The proposed system, called Dengue serotype NS1 Multiplex LFIA, provides high sensitivity and specificity. In testing for JEV, ZIKV, YFV, WNV, and CHIKV, the multiplex LFIA gave no indication of cross- reactivity with cell culture supernatants of other flaviviruses or chikungunya virus. In analyzing 187 samples from patients suspected of dengue infection, the detection sensitivity for serotype D1 to D4 was 90.0%, 88.24%, 82.61%, and 83.33% and serotype specificity was 98.74%, 96.13%, 99.39%, and 97.04%, respectively. Our multiplex LFIA can also identify mono- and co-infection of different serotype of dengue viruses in mosquitoes. The proposed Multiplex LFIA provides a simple tool for the rapid detection of dengue serotypes and in the differential diagnosis of fever patients in regions where medical resources are limited and/or multiple DENVs co-circulate.


Asunto(s)
Culicidae , Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales , Humanos , Inmunoensayo , Serogrupo , Proteínas no Estructurales Virales
6.
Front Immunol ; 12: 803807, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868089

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2021.761136.].

7.
Front Immunol ; 12: 761136, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34707621

RESUMEN

Scrub typhus (ST), also known as tsutsugamushi disease and caused by rickettsia Orientia tsutsugamushi, is an underestimated fatal epidemic in the Asia-Pacific region, resulting in a million human infections each year. ST is easily misdiagnosed as clinical diagnosis is based on non-specific skin eschar and flu-like symptoms. Thus, the lack of accurate, convenient, and low-cost detection methods for ST poses a global health threat. To address this problem, we adopted baculovirus surface-display technology to express three variants of TSA56, the major membrane antigen of O. tsutsugamushi, as well as the passenger domain of ScaC (ScaC-PD), on insect Sf21 cell surfaces rather than biosafety level 3 bacteria in an enzyme-linked immunosorbent assay (ELISA). Recombinant TSA56 and ScaC-PD were all properly expressed and displayed on Sf21 cells. Our cell-based ELISA comprising the four antigen-displaying cell types interacted with monoclonal antibodies as well as serum samples from ST-positive field-caught rats. This cell-based ELISA presented high accuracy (96.3%), sensitivity (98.6%), and specificity (84.6%) when tested against the ST-positive rat sera. Results of a pilot study using human sera were also highly consistent with the results of immunofluorescence analyses. By adopting this approach, we circumvented complex purification and refolding processes required to generate recombinant O. tsutsugamushi antigens and reduced the need for expensive equipment and extensively trained operators. Thus, our system has the potential to become a widely used serological platform for diagnosing ST.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Orientia tsutsugamushi/inmunología , Tifus por Ácaros/diagnóstico , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Baculoviridae/genética , Línea Celular , Técnicas de Visualización de Superficie Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Ratas , Proteínas Recombinantes/inmunología , Tifus por Ácaros/sangre , Tifus por Ácaros/inmunología , Spodoptera
8.
Sci Rep ; 11(1): 12321, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112897

RESUMEN

Reverse genetics is an important tool in the elucidation of viral replication and the development of countermeasures; however, these methods are impeded by laborious and inefficient replicon delivery methods. This paper demonstrates the use of a baculovirus to facilitate the efficient delivery of autonomous CHIKV replicons into mosquito and mammalian cells in vitro as well as adult mosquitoes in vivo. The efficacy of this approach was verified via co-localization among an eGFP reporter, nsP1, and dsRNA as well as through the inhibition of an RNA-dependent RNA polymerase (RdRp) null mutation (DDAA) in nsP4, or the treatment of a known antiviral compound (6-azauridine). We also investigated the correlation between CHIKV replicon-launched eGFP expression and the effectiveness of CHIKV replicon variants in inducing IFN-ß expression in human cell lines. This delivery method based on a single vector is applicable to mosquito and mammalian cells in seeking to decipher the mechanisms underlying CHIKV replication, elucidate virus-host interactions, and develop antivirals. This study presents an effective alternative to overcome many of the technological issues related to the study and utilization of autonomous arbovirus replicons.


Asunto(s)
Fiebre Chikungunya/genética , Virus Chikungunya/genética , ARN Polimerasa Dependiente del ARN/genética , Replicación Viral/genética , Aedes/virología , Animales , Antivirales/farmacología , Fiebre Chikungunya/transmisión , Fiebre Chikungunya/virología , Virus Chikungunya/patogenicidad , Chlorocebus aethiops/virología , Culicidae/virología , Humanos , Mosquitos Vectores/genética , Mosquitos Vectores/virología , ARN Viral/genética , Células Vero , Proteínas no Estructurales Virales/genética
9.
Int J Mol Sci ; 21(11)2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32517029

RESUMEN

Dengue virus (DENV) and Zika virus (ZIKV) are mosquito-borne flaviviruses that cause severe illness after infection. Currently, there are no specific or effective treatments against DENV and ZIKV. Previous studies have shown that tyrosine kinase activities and signal transduction are involved in flavivirus replication, suggesting a potential therapeutic strategy for DENV and ZIKV. In this study, we found that compound L3 can significantly reduce viral protein expression and viral titers in HEK-293, MCF-7, HepG2, and Huh-7 cells and exhibits superior therapeutic efficacy against flaviviral infection compared to other tyrosine kinase inhibitors. In addition, compound L3 can decrease endogenous HER2 activation and inhibit the phosphorylation of the HER2 downstream signaling molecules Src and ERK1/2, the levels of which have been associated with viral protein expression in MCF-7 cells. Moreover, silencing HER2 diminished DENV-2 and ZIKV expression in MCF-7 cells, which suggests that HER2 activity is involved in flavivirus replication. Furthermore, in DENV-2-infected AG129 mice, treatment with compound L3 increased the survival rates and reduced the viremia levels. Overall, compound L3 demonstrates therapeutic efficacy both in vitro and in vivo and could be developed as a promising antiviral drug against emerging flaviviruses or for concurrent DENV and ZIKV outbreaks.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus Zika/efectos de los fármacos , Afatinib/química , Afatinib/farmacología , Animales , Antivirales/química , Células Cultivadas , Dengue/virología , Relación Dosis-Respuesta a Droga , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Concentración 50 Inhibidora , Ratones , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/virología
10.
Viruses ; 12(3)2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32204533

RESUMEN

The Japanese encephalitis virus (JEV) is the major cause of an acute encephalitis syndrome in many Asian countries, despite the fact that an effective vaccine has been developed. Virus-like particles (VLPs) are self-assembled multi-subunit protein structures which possess specific epitope antigenicities related to corresponding native viruses. These properties mean that VLPs are considered safe antigens that can be used in clinical applications. In this study, we developed a novel baculovirus/mosquito (BacMos) expression system which potentially enables the scalable production of JEV genotype III (GIII) VLPs (which are secreted from mosquito cells). The mosquito-cell-derived JEV VLPs comprised 30-nm spherical particles as well as precursor membrane protein (prM) and envelope (E) proteins with densities that ranged from 30% to 55% across a sucrose gradient. We used IgM antibody-capture enzyme-linked immunosorbent assays to assess the resemblance between VLPs and authentic virions and thereby characterized the epitope specific antigenicity of VLPs. VLP immunization was found to elicit a specific immune response toward a balanced IgG2a/IgG1 ratio. This response effectively neutralized both JEV GI and GIII and elicited a mixed Th1/Th2 response in mice. This study supports the development of mosquito cell-derived JEV VLPs to serve as candidate vaccines against JEV.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Virus de la Encefalitis Japonesa (Especie)/ultraestructura , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/virología , Inmunidad Celular , Inmunidad Humoral , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Línea Celular , Culicidae/virología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Técnica del Anticuerpo Fluorescente , Ratones , Pruebas de Neutralización , Virión
11.
PLoS Pathog ; 15(8): e1007992, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31381617

RESUMEN

Genotype I (GI) virus has replaced genotype III (GIII) virus as the dominant Japanese encephalitis virus (JEV) in the epidemic area of Asia. The mechanism underlying the genotype replacement remains unclear. Therefore, we focused our current study on investigating the roles of mosquito vector and amplifying host(s) in JEV genotype replacement by comparing the replication ability of GI and GIII viruses. GI and GIII viruses had similar infection rates and replicated to similar viral titers after blood meal feedings in Culex tritaeniorhynchus. However, GI virus yielded a higher viral titer in amplifying host-derived cells, especially at an elevated temperature, and produced an earlier and higher viremia in experimentally inoculated pigs, ducklings, and young chickens. Subsequently we identified the amplification advantage of viral genetic determinants from GI viruses by utilizing chimeric and recombinant JEVs (rJEVs). Compared to the recombinant GIII virus (rGIII virus), we observed that both the recombinant GI virus and the chimeric rJEVs encoding GI virus-derived NS1-3 genes supported higher replication ability in amplifying hosts. The replication advantage of the chimeric rJEVs was lost after introduction of a single substitution from a GIII viral mutation (NS2B-L99V, NS3-S78A, or NS3-D177E). In addition, the gain-of-function assay further elucidated that rGIII virus encoding GI virus NS2B-V99L/NS3-A78S/E177E substitutions re-gained the enhanced replication ability. Thus, we conclude that the replication advantage of GI virus in pigs and poultry is the result of three critical NS2B/NS3 substitutions. This may lead to more efficient transmission of GI virus than GIII virus in the amplifying host-mosquito cycle.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/virología , Mosquitos Vectores , Mutación , Proteínas no Estructurales Virales/genética , Viremia/transmisión , Animales , Pollos , Culex , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Encefalitis Japonesa/epidemiología , Encefalitis Japonesa/genética , Femenino , Genotipo , ARN Helicasas/genética , Serina Endopeptidasas/genética , Porcinos , Replicación Viral
12.
J Clin Microbiol ; 57(7)2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30971466

RESUMEN

Dengue fever, caused by infections with the dengue virus (DENV), affects nearly 400 million people globally every year. Early diagnosis and management can reduce the morbidity and mortality rates of severe forms of dengue disease as well as decrease the risk of wider outbreaks. Although the early diagnosis of dengue can be achieved using a number of commercial NS1 detection kits, none of these can differentiate among the four dengue virus serotypes. In this study, we developed an enzyme-linked immunosorbent assay (ELISA) for the detection of dengue virus (DENV) NS1 by pairing a serotype-cross-reactive monoclonal antibody (MAb) with one of four serotype-specific MAbs in order to facilitate the rapid detection of NS1 antigens and the simultaneous differentiation of DENV serotypes. A total of 146 serum samples obtained from patients suspected to be in the acute phase of DENV infection were used to evaluate the clinical application of our novel test for the detection and serotyping of DENV. The overall sensitivity rate of our test was 84.85%, and the sensitivity rates for serotyping were as follows: 88.2% (15/17) for DENV serotype 1 (DENV1), 94.7% (18/19) for DENV2, 75% (12/16) for DENV3, and 66.6% (6/9) for DENV4. Moreover, there was no cross-reactivity among serotypes, and no cross-reactivity was observed in sera from nondengue patients. Thus, our test not only enables the rapid detection of the dengue virus but also can distinguish among the specific serotypes during the early stages of infection. These results indicate that our ELISA for DENV NS1 is a convenient tool that may help elucidate the epidemiology of DENV outbreaks and facilitate the clinical management of DENV infections.


Asunto(s)
Antígenos Virales/sangre , Técnicas de Laboratorio Clínico/métodos , Virus del Dengue/aislamiento & purificación , Dengue/diagnóstico , Proteínas no Estructurales Virales/sangre , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Dengue/sangre , Virus del Dengue/clasificación , Virus del Dengue/inmunología , Diagnóstico Precoz , Ensayo de Inmunoadsorción Enzimática , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Serogrupo , Serotipificación
13.
Sci Rep ; 8(1): 17778, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30542209

RESUMEN

Efficient gene delivery technologies play an essential role in the gene functional analyses that are necessary for basic and applied researches. Mosquitoes are ubiquitous insects, responsible for transmitting many deadly arboviruses causing millions of human deaths every year. The lack of efficient and flexible gene delivery strategies in mosquitoes are among the major hurdles for the study of mosquito biology and mosquito-pathogen interactions. We found that Autographa californica multiple nucleopolyhedrovirus (AcMNPV), the type baculovirus species, can efficiently transduce mosquito cells without viral propagation, allowing high level gene expression upon inducement by suitable promoters without obvious negative effects on cell propagation and viability. AcMNPV transduces into several mosquito cell types, efficiently than in commonly used mammalian cell lines and classical plasmid DNA transfection approaches. We demonstrated the application of this system by expressing influenza virus neuraminidase (NA) into mosquito hosts. Moreover, AcMNPV can transduce both larvae and adults of essentially all blood-sucking mosquito genera, resulting in bright fluorescence in insect bodies with little or no tissue barriers. Our experiments establish baculovirus as a convenient and powerful gene delivery vector in vitro and in vivo that will greatly benefit research into mosquito gene regulation, development and the study of mosquito-borne viruses.


Asunto(s)
Baculoviridae/genética , Culicidae/genética , Culicidae/virología , Mosquitos Vectores/genética , Animales , Línea Celular , Chlorocebus aethiops , Vectores de Enfermedades , Expresión Génica/genética , Técnicas de Transferencia de Gen , Células HEK293 , Humanos , Larva/genética , Larva/virología , Neuraminidasa/genética , Nucleopoliedrovirus/genética , Orthomyxoviridae/genética , Transfección/métodos , Células Vero
14.
Antiviral Res ; 159: 134-142, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30300716

RESUMEN

The chikungunya virus (CHIKV) is a mosquito-borne virus that belongs to the genus Alphavirus, family Togaviridae. It is the cause of chikungunya fever in humans, which presents a serious global threat due to its high rate of contagion. The clinical symptoms of CHIKV include fever and persistent, severe arthritis. Micafungin has broad-spectrum fungicidal activity against Candida spp. is a promising echinocandin that was recently approved by the U.S. Food and Drug Administration (FDA) and has demonstrated activity against Candida and Aspergillus. Recent studies have demonstrated the antiviral activity of micafungin; however, the inhibitory effects against CHIKV have yet to be investigated. Our objectives in this study were to explore the antiviral effects of micafungin on CHIKV infection and to elucidate the potential molecular mechanisms of inhibition. We determined that micafungin has the ability to counter CHIKV-induced cytopathic effects. We further discovered that micafungin limits virus replication, release, cell-to-cell transmission, and also slightly affected virus stability during high doses treatment. The efficacy of micafungin was further confirmed against two clinical isolates of CHIKV and two alphaviruses: Sindbis virus (SINV) and Semliki Forest virus (SFV). Our findings suggest that micafungin has considerable potential as a novel inhibitor against the viral replication, and intracellular and extracellular transmission of CHIKV, and has a little effect on virus stability. Our findings also suggest that micafungin could have curative effects on other alphavirus infections.


Asunto(s)
Alphavirus/efectos de los fármacos , Antivirales/farmacología , Virus Chikungunya/efectos de los fármacos , Micafungina/farmacología , Infecciones por Alphavirus/tratamiento farmacológico , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/virología , Virus de los Bosques Semliki/efectos de los fármacos , Virus Sindbis/efectos de los fármacos , Replicación Viral/efectos de los fármacos
15.
Biochem Biophys Res Commun ; 491(3): 595-602, 2017 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-28760340

RESUMEN

Chikungunya is a severe disease that results from infection with the chikungunya virus (CHIKV), an arbovirus. Thus, we (1) explored a new approach to combining previously researched drugs that have shown the potential to inhibit CHIKV infection; and (2) demonstrated the antiviral effects of (-)-Epigallocatechin-3-gallate (EGCG) and the underlying mechanisms. Specifically, we used U2OS cells infected with CHIVK to assess the synergistic antiviral activities of EGCG and suramin. EGCG presented the ability to inhibit the viral RNA, progeny yield, and cytopathic effect (CPE) of CHIKV and also demonstrated the ability to protect against virus entry, replication, and release. Moreover, the results confirmed that EGCG and suramin can have synergistic effects against CHIKV strain S27 infection and two other clinical isolates of CHIKV. Our findings suggest that treatment with a combination of EGCG and suramin could provide a basis for the development of novel stretages against CHIKV infection.


Asunto(s)
Catequina/análogos & derivados , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/virología , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/fisiología , Suramina/administración & dosificación , Antivirales/administración & dosificación , Catequina/administración & dosificación , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Sinergismo Farmacológico , Humanos , Resultado del Tratamiento
16.
Antiviral Res ; 135: 81-90, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27742486

RESUMEN

Chikungunya disease results from an infection with the arbovirus, chikungunya virus (CHIKV). Symptoms of CHIKV include fever and persistent, severe arthritis. In recent years, several antiviral drugs have been evaluated in clinical trials; however, no registered antivirals have been approved for clinical therapy. In this study, we established a high-throughput screening (HTS) system based on CHIKV 26S mediated insect cell fusion inhibition assay. Our screening system was able to search potential anti-CHIKV drugs in vitro. Using this system, four compounds (niclosamide, nitazoxanide, niflumic acid, tolfenamic acid) were identified. These compounds were then further analyzed using a microneutralization assay. We determined that niclosamide and nitazoxanide exhibit ability to against CHIKV-induced CPE. The anti-CHIKV abilities of these compounds were further confirmed by RT-qPCR and IFA. Moreover, niclosamide and nitazoxanide were found to (1) limit virus entry, (2) inhibit both viral release and cell-to-cell transmission, and (3) possess broad anti-alphavius activities, including against two clinical CHIKV isolates and two alphaviruses: Sindbis virus (SINV) and Semliki forest virus (SFV). In conclusion, our findings suggested that niclosamide and nitazoxanide were able to inhibit CHIKV entry and transmission, which might provide a basis for the development of novel human drug therapies against CHIKV and other alphavirus infections.


Asunto(s)
Antivirales/farmacología , Virus Chikungunya/efectos de los fármacos , Descubrimiento de Drogas , Niclosamida/farmacología , Tiazoles/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Línea Celular , Fiebre Chikungunya/transmisión , Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Ensayos Analíticos de Alto Rendimiento , Humanos , Nitrocompuestos , Virus de los Bosques Semliki/efectos de los fármacos , Virus Sindbis/efectos de los fármacos , Replicación Viral/efectos de los fármacos
17.
Antiviral Res ; 134: 89-96, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27577529

RESUMEN

The chikungunya virus (CHIKV), an arthritogenic alphavirus, has caused explosive epidemics involving millions of cases. Globally expanding pandemics involving CHIKV and post-CHIKV rheumatic disorders are increasing public health concerns. However, no antiviral interventions or vaccines to control CHIKV infection have yet been approved. Although suramin has been possess anti-CHIKV activity in vitro, whether suramin has anti-CHIKV activity in vivo remains unknown. This study aimed to determine whether suramin treatment could ameliorate CHIKV-induced arthritis in a C57BL/6 mice model. C57BL/6 mice were infected with CHIKVs to evaluate anti-CHIKV activities of suramin in terms of histopathology, viral burden and disease score. Not only did suramin treatment substantially decrease viral loads, but it also significantly ameliorated acute foot lesions in mice. In addition, suramin treatment markedly restores cartilage integrity and reduces the number of IHC positive chondrocyte in mice infected with CHIKV strains 0810bTw and 0706aTw. This in vivo study highlights the potential ability of suramin to treat CHIKV infection in clinical settings.


Asunto(s)
Antivirales/uso terapéutico , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/efectos de los fármacos , Suramina/uso terapéutico , Animales , Virus Chikungunya/patogenicidad , Modelos Animales de Enfermedad , Pie/patología , Pie/virología , Ratones , Ratones Endogámicos C57BL , Enfermedades Musculoesqueléticas/tratamiento farmacológico , Enfermedades Musculoesqueléticas/etiología , Enfermedades Musculoesqueléticas/virología , Suramina/administración & dosificación , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
18.
PLoS One ; 10(7): e0133511, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26208101

RESUMEN

The mosquito-borne Chikungunya virus (CHIKV) is a profound global threat due to its high rate of contagion and the lack of vaccine or effective treatment. Suramin is a symmetric polyanionic naphthylurea that is widely used in the clinical treatment of parasite infections. Numerous studies have reported the broad antiviral activities of suramin; however, inhibition effects against CHIKV have not yet been demonstrated. The aim of this study was thus to investigate the antiviral effect of suramin on CHIKV infection and to elucidate the molecular mechanism underlying inhibition using plaque reduction assay, RT-qPCR, western blot analysis, and plaque assay. Microneutralization assay was used to determine the EC50 of suramin in the CHIKV-S27 strain as well as in three other clinical strains (0611aTw, 0810bTw and 0706aTw). Time-of-addition was used to reveal the anti-CHIKV mechanism of suramin. We also evaluated anti-CHIKV activity with regard to viral entry, virus release, and cell-to-cell transmission. Cytopathic effect, viral RNA, viral protein, and the virus yield of CHIKV infection were shown to diminish in the presence of suramin in a dose-dependent manner. Suramin was also shown the inhibitory activities of the three clinical isolates. Suramin inhibited the early progression of CHIKV infection, due perhaps to interference with virus fusion and binding, which subsequently prevented viral entry. Results of a molecular docking simulation indicate that suramin may embed within the cavity of the E1/E2 heterodimer to interfere with their function. Suramin was also shown to reduce viral release and cell-to-cell transmission of CHIKV. In conclusion, Suramin shows considerable potential as a novel anti-CHIKV agent targeting viral entry, extracellular transmission, and cell-to-cell transmission.


Asunto(s)
Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Suramina/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/química , Antivirales/farmacología , Línea Celular , Modelos Moleculares , Conformación Molecular , Suramina/química , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Ensayo de Placa Viral , Liberación del Virus/efectos de los fármacos
19.
Biochem Biophys Res Commun ; 396(3): 596-601, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20412785

RESUMEN

microRNAs (miRNAs) are endogenous non-coding RNAs that spatiotemporally modulate mRNAs in a post-transcriptional manner. The engineering of viruses by insertion of a tissue-specific miRNA recognition element (MRE) into viral mRNA can restrict viral tissue tropism. In this study we employed dengue virus (DEN) replicons to investigate whether miRNAs are able to suppress flavivirus replication through the targeting of non-polyadenylated viral mRNA. Because liver infection by DEN may contribute to the virus pathogenesis, we inserted an MRE of hepatic-specific microRNA-122 (miR-122) into its 3'-untranslated region (3'-UTR) to test the feasibility of creating a liver-restricted DEN replicon. Our results demonstrate that incorporation of the miR-122-MRE confers upon the DEN replicon an inhibitory susceptibility to miR-122 targeting, suggesting that DEN can be engineered to exert the desired replication restriction effect to avoid infection of vital tissues/organs. This approach provides an additional layer of biosafety and thus has great potential for use in the rational development of safer flavivirus vaccines.


Asunto(s)
Virus del Dengue/fisiología , Regulación Viral de la Expresión Génica , Ingeniería Genética/métodos , Hígado/virología , MicroARNs/genética , Replicón , Replicación Viral , Regiones no Traducidas 3'/genética , Animales , Línea Celular Tumoral , Cricetinae , Virus del Dengue/genética , Humanos
20.
J Biol Chem ; 279(5): 3308-17, 2004 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-14607839

RESUMEN

Two mosquito STATs, AaSTAT and CtSTAT, have been cloned from Aedes albopictus and Culex tritaeniorhynchus mosquitoes, respectively. These two STATs are more similar to those of Drosophila, Anopheles, and mammalian STAT5 in the DNA binding and Src homology 2 domains. The mRNA transcripts are expressed at all developmental stages, and the proteins are present predominantly at the pupal and adult stages in both mosquitoes. Stimulation with lipopolysaccharide resulted in an increase of tyrosine phosphorylation and DNA binding activity of AaSTAT and CtSTAT as well as an increase of luciferase activity of a reporter gene containing Drosophila STAT binding motif in mosquito C6/36 cells. After being infected with Japanese encephalitis virus, nuclear extracts of C6/36 cells revealed a decrease of tyrosine phosphorylation and DNA binding activity of AaSTAT which could be restored by sodium orthovanadate treatment. Taking all of the data together, this is the first report to clone and characterize two mosquito STATs with 81% identity and to demonstrate a different response of tyrosine phosphorylation and DNA binding of these two STATs by lipopolysaccharide treatment and by Japanese encephalitis virus infection.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Proteínas de Insectos , Transducción de Señal , Transactivadores/química , Tirosina/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Culicidae/virología , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Drosophila , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Lipopolisacáridos/química , Masculino , Datos de Secuencia Molecular , Fosforilación , Filogenia , Plásmidos/metabolismo , Unión Proteica , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Factores de Transcripción STAT , Homología de Secuencia de Aminoácido , Factores Sexuales , Factores de Tiempo , Distribución Tisular , Transcripción Genética , Activación Transcripcional , Vanadatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...