Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.788
Filtrar
1.
Cells ; 13(10)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38786028

RESUMEN

Zika (ZIKV) and Chikungunya (CHIKV) viruses are mosquito-transmitted infections, or vector-borne pathogens, that emerged a few years ago. Reliable diagnostic tools for ZIKV and CHIKV-inexpensive, multiplexed, rapid, highly sensitive, and specific point-of-care (POC) systems-are vital for appropriate risk management and therapy. We recently studied a detection system with great success in Mexico (Villahermosa, state of Tabasco), working with human sera from patients infected with those viruses. The research conducted in Mexico validated the efficacy of a novel two-step rapid isothermal amplification technique (RAMP). This approach, which encompasses recombinase polymerase amplification (RPA) followed by loop-mediated isothermal amplification (LAMP), had been previously established in the lab using lab-derived Zika (ZIKV) and Chikungunya (CHIKV) viruses. Crucially, our findings confirmed that this technique is also effective when applied to human sera samples collected from locally infected individuals in Mexico.


Asunto(s)
Virus Chikungunya , Técnicas de Amplificación de Ácido Nucleico , Infección por el Virus Zika , Virus Zika , Humanos , Virus Zika/genética , Virus Zika/aislamiento & purificación , Técnicas de Amplificación de Ácido Nucleico/métodos , Virus Chikungunya/genética , Virus Chikungunya/aislamiento & purificación , Infección por el Virus Zika/diagnóstico , Infección por el Virus Zika/virología , Infección por el Virus Zika/sangre , Fiebre Chikungunya/diagnóstico , Fiebre Chikungunya/virología , Fiebre Chikungunya/sangre , Técnicas de Diagnóstico Molecular/métodos , ARN Viral/genética , ARN Viral/sangre , México , Sensibilidad y Especificidad , Virus ARN/genética , Virus ARN/aislamiento & purificación
2.
Proc Natl Acad Sci U S A ; 121(21): e2312755121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38743628

RESUMEN

Antigenic similarities between Zika virus (ZIKV) and other flaviviruses pose challenges to the development of virus-specific diagnostic tools and effective vaccines. Starting with a DNA-encoded one-bead-one-compound combinatorial library of 508,032 synthetic, non-natural oligomers, we selected and characterized small molecules that mimic ZIKV epitopes. High-throughput fluorescence-activated cell sorter-based bead screening was used to select molecules that bound IgG from ZIKV-immune but not from dengue-immune sera. Deep sequencing of the DNA from the "Zika-only" beads identified 40 candidate molecular structures. A lead candidate small molecule "CZV1-1" was selected that correctly identifies serum specimens from Zika-experienced patients with good sensitivity and specificity (85.3% and 98.4%, respectively). Binding competition studies of purified anti-CZV1-1 IgG against known ZIKV-specific monoclonal antibodies (mAbs) showed that CZV1-1 mimics a nonlinear, neutralizing conformational epitope in the domain III of the ZIKV envelope. Purified anti-CZV1-1 IgG neutralized infection of ZIKV in cell cultures with potencies comparable to highly specific ZIKV-neutralizing mAbs. This study demonstrates an innovative approach for identification of synthetic non-natural molecular mimics of conformational virus epitopes. Such molecular mimics may have value in the development of accurate diagnostic assays for Zika, as well as for other viruses.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Epítopos , Infección por el Virus Zika , Virus Zika , Virus Zika/inmunología , Epítopos/inmunología , Humanos , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Inmunoglobulina G/inmunología , Anticuerpos Monoclonales/inmunología , Imitación Molecular/inmunología
3.
Sci Rep ; 14(1): 10407, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710792

RESUMEN

Glucose regulated protein 78 (GRP78) is a chaperone protein that is a central mediator of the unfolded protein response, a key cellular stress response pathway. GRP78 has been shown to be critically required for infection and replication of a number of flaviviruses, and to interact with both non-structural (NS) and structural flavivirus proteins. However, the nature of the specific interaction between GRP78 and viral proteins remains largely unknown. This study aimed to characterize the binding domain and critical amino acid residues that mediate the interaction of GRP78 to ZIKV E and NS1 proteins. Recombinant EGFP fused GRP78 and individual subdomains (the nucleotide binding domain (NBD) and the substrate binding domain (SBD)) were used as a bait protein and co-expressed with full length or truncated ZIKV E and NS1 proteins in HEK293T/17 cells. Protein-protein interactions were determined by a co-immunoprecipitation assay. From the results, both the NBD and the SBD of GRP78 were crucial for an effective interaction. Single amino acid substitutions in the SBD showed that R492E and T518A mutants significantly reduced the binding affinity of GRP78 to ZIKV E and NS1 proteins. Notably, the interaction of GRP78 with ZIKV E was stably maintained against various single amino acid substitutions on ZIKV E domain III and with all truncated ZIKV E and NS1 proteins. Collectively, the results suggest that the principal binding between GRP78 and viral proteins is mainly a classic canonical chaperone protein-client interaction. The blocking of GRP78 chaperone function effectively inhibited ZIKV infection and replication in neuronal progenitor cells. Our findings reveal that GRP78 is a potential host target for anti-ZIKV therapeutics.


Asunto(s)
Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico , Unión Proteica , Proteínas no Estructurales Virales , Virus Zika , Chaperón BiP del Retículo Endoplásmico/metabolismo , Virus Zika/metabolismo , Virus Zika/fisiología , Humanos , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Células HEK293 , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología , Replicación Viral
4.
PLoS One ; 19(5): e0281851, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38748732

RESUMEN

Zika (ZIKV) and chikungunya (CHIKV) are arboviruses that cause infections in humans and can cause clinical complications, representing a worldwide public health problem. Aedes aegypti is the primary vector of these pathogens and Culex quinquefasciatus may be a potential ZIKV vector. This study aimed to evaluate fecundity, fertility, survival, longevity, and blood feeding activity in Ae. aegypti after exposure to ZIKV and CHIKV and, in Cx. quinquefasciatus exposed to ZIKV. Three colonies were evaluated: AeCamp (Ae. aegypti-field), RecL (Ae. aegypti-laboratory) and CqSLab (Cx. quinquefasciatus-laboratory). Seven to 10 days-old females from these colonies were exposed to artificial blood feeding with CHIKV or ZIKV. CHIKV caused reduction in fecundity and fertility in AeCamp and reduction in survival and fertility in RecL. ZIKV impacted survival in RecL, fertility in AeCamp and, fecundity and fertility in CqSLab. Both viruses had no effect on blood feeding activity. These results show that CHIKV produces a higher biological cost in Ae. aegypti, compared to ZIKV, and ZIKV differently alters the biological performance in colonies of Ae. aegypti and Cx. quinquefasciatus. These results provide a better understanding over the processes of virus-vector interaction and can shed light on the complexity of arbovirus transmission.


Asunto(s)
Aedes , Virus Chikungunya , Culex , Fertilidad , Mosquitos Vectores , Infección por el Virus Zika , Virus Zika , Animales , Aedes/virología , Aedes/fisiología , Virus Chikungunya/fisiología , Virus Chikungunya/patogenicidad , Virus Zika/fisiología , Virus Zika/patogenicidad , Culex/virología , Culex/fisiología , Mosquitos Vectores/virología , Mosquitos Vectores/fisiología , Femenino , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología , Fiebre Chikungunya/transmisión , Fiebre Chikungunya/virología , Conducta Alimentaria/fisiología , Humanos , Longevidad
5.
Viruses ; 16(5)2024 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-38793688

RESUMEN

Arboviral diseases are serious threats to global health with increasing prevalence and potentially severe complications. Significant arthropod-borne viruses are the dengue viruses (DENV 1-4), the Zika virus (ZIKV), and the chikungunya virus (CHIKV). Among the areas most affected is the South Pacific Region (SPR). Here, arboviruses not only cause a high local burden of disease, but the region has also proven to contribute to their global spread. Outpatient serum samples collected between 08/2016 and 04/2017 on three islands of the island states of Vanuatu and the Cook Islands were tested for anti-DENV- and anti-ZIKV-specific antibodies (IgG) using enzyme-linked immunosorbent assays (ELISA). ELISA test results showed 89% of all test sera from the Cook Islands and 85% of the Vanuatu samples to be positive for anti-DENV-specific antibodies. Anti-ZIKV antibodies were identified in 66% and 52%, respectively, of the test populations. Statistically significant differences in standardized immunity levels were found only at the intranational level. Our results show that in both the Cook Islands and Vanuatu, residents were exposed to significant Flavivirus transmission. Compared to other seroprevalence studies, the marked difference between ZIKV immunity levels and previously published CHIKV seroprevalence rates in our study populations is surprising. We propose the timing of ZIKV and CHIKV emergence in relation to recurrent DENV outbreaks and the impact of seasonality as explanatory external factors for this observation. Our data add to the knowledge of arboviral epidemics in the SPR and contribute to a better understanding of virus spread, including external conditions with potential influence on outbreak dynamics. These data may support preventive and rapid response measures in the affected areas, travel-related risk assessment, and infection identification in locals and returning travelers.


Asunto(s)
Anticuerpos Antivirales , Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Humanos , Infección por el Virus Zika/epidemiología , Infección por el Virus Zika/sangre , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Estudios Seroepidemiológicos , Virus del Dengue/inmunología , Virus Zika/inmunología , Vanuatu/epidemiología , Dengue/epidemiología , Dengue/inmunología , Dengue/sangre , Dengue/virología , Polinesia/epidemiología , Anticuerpos Antivirales/sangre , Adulto , Femenino , Adolescente , Adulto Joven , Masculino , Persona de Mediana Edad , Anciano , Niño , Ensayo de Inmunoadsorción Enzimática , Preescolar , Inmunoglobulina G/sangre , Lactante
6.
Sci Signal ; 17(837): eadi9844, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38771918

RESUMEN

Oligoadenylate synthetase 3 (OAS3) and ribonuclease L (RNase L) are components of a pathway that combats viral infection in mammals. Upon detection of viral double-stranded RNA (dsRNA), OAS3 synthesizes 2'-5'-oligo(A), which activates the RNase domain of RNase L by promoting the homodimerization and oligomerization of RNase L monomers. Activated RNase L rapidly degrades all cellular mRNAs, shutting off several cellular processes. We sought to understand the molecular mechanisms underlying the rapid activation of RNase L in response to viral infection. Through superresolution microscopy and live-cell imaging, we showed that OAS3 and RNase L concentrated into higher-order cytoplasmic complexes known as dsRNA-induced foci (dRIF) in response to dsRNA or infection with dengue virus, Zika virus, or West Nile virus. The concentration of OAS3 and RNase L at dRIF corresponded with the activation of RNase L-mediated RNA decay. We showed that dimerized/oligomerized RNase L concentrated in a liquid-like shell surrounding a core OAS3-dRIF structure and dynamically exchanged with the cytosol. These data establish that the condensation of dsRNA, OAS3, and RNase L into dRIF is a molecular switch that promotes the rapid activation of RNase L upon detection of dsRNA in mammalian cells.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Endorribonucleasas , ARN Bicatenario , Virus Zika , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/química , Humanos , 2',5'-Oligoadenilato Sintetasa/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/química , ARN Bicatenario/metabolismo , ARN Bicatenario/química , ARN Bicatenario/genética , Virus Zika/metabolismo , Animales , Virus del Dengue/metabolismo , ARN Viral/metabolismo , ARN Viral/genética , Estabilidad del ARN , Virus del Nilo Occidental/metabolismo , Virus del Nilo Occidental/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología , Activación Enzimática , Células HeLa , Células HEK293
7.
Sci Transl Med ; 16(749): eadn2199, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38809964

RESUMEN

Infection with any of the four dengue virus serotypes (DENV1-4) can protect against or enhance subsequent dengue depending on preexisting antibodies and infecting serotype. Additionally, primary infection with the related flavivirus Zika virus (ZIKV) is associated with increased risk of DENV2 disease. Here, we measured how prior DENV and ZIKV immunity influenced risk of disease caused by DENV1-4 in a pediatric Nicaraguan cohort. Of 3412 participants in 2022, 10.6% experienced dengue cases caused by DENV1 (n = 139), DENV4 (n = 133), DENV3 (n = 54), DENV2 (n = 9), or an undetermined serotype (n = 39). Longitudinal clinical and serological data were used to define infection histories, and generalized linear and additive models adjusted for age, sex, time since last infection, and year, and repeat measurements were used to predict disease risk. Compared with flavivirus-naïve participants, primary ZIKV infection was associated with increased risk of disease caused by DENV4 (relative risk = 2.62, 95% confidence interval: 1.48 to 4.63) and DENV3 (2.90, 1.34 to 6.27), but not DENV1 infection. Primary DENV infection or DENV followed by ZIKV infection was also associated with increased risk of DENV4 disease. We reanalyzed 19 years of cohort data and demonstrated that prior flavivirus immunity and antibody titer had distinct associations with disease risk depending on incoming serotype. We thus find that prior ZIKV infection, like prior DENV infection, is associated with increased risk of disease with certain DENV serotypes. Cross-reactivity among flaviviruses should be considered when assessing vaccine safety and efficacy.


Asunto(s)
Virus del Dengue , Dengue , Serogrupo , Infección por el Virus Zika , Virus Zika , Humanos , Virus Zika/inmunología , Dengue/inmunología , Dengue/virología , Virus del Dengue/inmunología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Niño , Femenino , Masculino , Nicaragua/epidemiología , Preescolar , Factores de Riesgo , Adolescente , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Estudios de Cohortes
8.
Science ; 384(6693): eadn9524, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38669573

RESUMEN

The commensal microbiota of the mosquito gut plays a complex role in determining the vector competence for arboviruses. In this study, we identified a bacterium from the gut of field Aedes albopictus mosquitoes named Rosenbergiella sp. YN46 (Rosenbergiella_YN46) that rendered mosquitoes refractory to infection with dengue and Zika viruses. Inoculation of 1.6 × 103 colony forming units (CFUs) of Rosenbergiella_YN46 into A. albopictus mosquitoes effectively prevents viral infection. Mechanistically, this bacterium secretes glucose dehydrogenase (RyGDH), which acidifies the gut lumen of fed mosquitoes, causing irreversible conformational changes in the flavivirus envelope protein that prevent viral entry into cells. In semifield conditions, Rosenbergiella_YN46 exhibits effective transstadial transmission in field mosquitoes, which blocks transmission of dengue virus by newly emerged adult mosquitoes. The prevalence of Rosenbergiella_YN46 is greater in mosquitoes from low-dengue areas (52.9 to ~91.7%) than in those from dengue-endemic regions (0 to ~6.7%). Rosenbergiella_YN46 may offer an effective and safe lead for flavivirus biocontrol.


Asunto(s)
Aedes , Virus del Dengue , Mosquitos Vectores , Simbiosis , Virus Zika , Animales , Aedes/microbiología , Aedes/virología , Virus del Dengue/fisiología , Mosquitos Vectores/virología , Mosquitos Vectores/microbiología , Virus Zika/fisiología , Dengue/transmisión , Dengue/virología , Dengue/prevención & control , Microbioma Gastrointestinal , Acetobacteraceae/fisiología , Femenino , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Flavivirus/fisiología , Flavivirus/genética , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología
11.
Emerg Microbes Infect ; 13(1): 2348528, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38662785

RESUMEN

Zika is a systemic inflammatory disease caused by infection with Zika virus (ZIKV). ZIKV infection in adults is associated with encephalitis marked by elevated expression of pro-inflammatory cytokines and chemokines, as well as increased brain infiltration of immune cells. In this study, we demonstrate that ZIKV encephalitis in a mouse infection model exhibits increased brain TSPO expression. TSPO expression on brain-resident and infiltrating immune cells in ZIKV infection correlates with disease and inflammation status in the brain. Brain TSPO expression can also be sensitively detected ex vivo and in vitro using radioactive small molecule probes that specifically bind to TSPO, such as [3H]PK11195. TSPO expression on brain-resident and infiltrating immune cells is a biomarker of ZIKV neuroinflammation, which can also be a general biomarker of acute viral neuroinflammatory disease.


Asunto(s)
Biomarcadores , Encéfalo , Enfermedades Neuroinflamatorias , Receptores de GABA , Infección por el Virus Zika , Virus Zika , Animales , Infección por el Virus Zika/virología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/metabolismo , Ratones , Receptores de GABA/metabolismo , Receptores de GABA/genética , Virus Zika/inmunología , Encéfalo/virología , Encéfalo/metabolismo , Encéfalo/patología , Enfermedades Neuroinflamatorias/virología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Femenino , Citocinas/metabolismo
12.
Viruses ; 16(4)2024 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-38675882

RESUMEN

As a mosquito-borne flavivirus, Zika virus (ZIKV) has been identified as a global health threat. The virus has been linked to severe congenital disabilities, including microcephaly and other congenital malformations, resulting in fatal intrauterine death. Therefore, developing sensitive and specific methods for the early detection and accurate diagnosis of the ZIKV is essential for controlling its spread and mitigating its impact on public health. Herein, we set up a novel nucleic acid detection system based on Pyrococcus furiosus Argonaute (PfAgo)-mediated nucleic acid detection, targeting the non-structural protein 5 (NS5) region of the ZIKV genome (abbreviated ZIKV-PAND). Without preamplification with the polymerase chain reaction (PCR), the minimum detection concentration (MDC) of ZIKV-PAND was about 10 nM. When introducing an amplification step, the MDC can be dramatically decreased to the aM level (8.3 aM), which is comparable to qRT-PCR assay (1.6 aM). In addition, the diagnostic findings from the analysis of simulated clinical samples or Zika virus samples using ZIKV-PAND show a complete agreement of 100% with qRT-PCR assays. This correlation can aid in the implementation of molecular testing for clinical diagnoses and the investigation of ZIKV infection on an epidemiological scale.


Asunto(s)
Pyrococcus furiosus , Proteínas no Estructurales Virales , Infección por el Virus Zika , Virus Zika , Virus Zika/genética , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/diagnóstico , Infección por el Virus Zika/virología , Humanos , Proteínas no Estructurales Virales/genética , Pyrococcus furiosus/genética , Proteínas Argonautas/genética , Sensibilidad y Especificidad , ARN Viral/genética , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Amplificación de Ácido Nucleico/métodos , Genoma Viral
13.
J Virol ; 98(5): e0019424, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38567950

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus that caused an epidemic in the Americas in 2016 and is linked to severe neonatal birth defects, including microcephaly and spontaneous abortion. To better understand the host response to ZIKV infection, we adapted the 10× Genomics Chromium single-cell RNA sequencing (scRNA-seq) assay to simultaneously capture viral RNA and host mRNA. Using this assay, we profiled the antiviral landscape in a population of human monocyte-derived dendritic cells infected with ZIKV at the single-cell level. The bystander cells, which lacked detectable viral RNA, expressed an antiviral state that was enriched for genes coinciding predominantly with a type I interferon (IFN) response. Within the infected cells, viral RNA negatively correlated with type I IFN-dependent and -independent genes (the antiviral module). We modeled the ZIKV-specific antiviral state at the protein level, leveraging experimentally derived protein interaction data. We identified a highly interconnected network between the antiviral module and other host proteins. In this work, we propose a new paradigm for evaluating the antiviral response to a specific virus, combining an unbiased list of genes that highly correlate with viral RNA on a per-cell basis with experimental protein interaction data. IMPORTANCE: Zika virus (ZIKV) remains a public health threat given its potential for re-emergence and the detrimental fetal outcomes associated with infection during pregnancy. Understanding the dynamics between ZIKV and its host is critical to understanding ZIKV pathogenesis. Through ZIKV-inclusive single-cell RNA sequencing (scRNA-seq), we demonstrate on the single-cell level the dynamic interplay between ZIKV and the host: the transcriptional program that restricts viral infection and ZIKV-mediated inhibition of that response. Our ZIKV-inclusive scRNA-seq assay will serve as a useful tool for gaining greater insight into the host response to ZIKV and can be applied more broadly to the flavivirus field.


Asunto(s)
Células Dendríticas , Análisis de la Célula Individual , Infección por el Virus Zika , Virus Zika , Humanos , Virus Zika/fisiología , Infección por el Virus Zika/virología , Infección por el Virus Zika/inmunología , Células Dendríticas/virología , Células Dendríticas/inmunología , ARN Viral/metabolismo , ARN Viral/genética , Interferón Tipo I/metabolismo , Interacciones Huésped-Patógeno , Análisis de Secuencia de ARN
14.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673962

RESUMEN

In the global pandemic scenario, dengue and zika viruses (DENV and ZIKV, respectively), both mosquito-borne members of the flaviviridae family, represent a serious health problem, and considering the absence of specific antiviral drugs and available vaccines, there is a dire need to identify new targets to treat these types of viral infections. Within this drug discovery process, the protease NS2B/NS3 is considered the primary target for the development of novel anti-flavivirus drugs. The NS2B/NS3 is a serine protease that has a dual function both in the viral replication process and in the elusion of the innate immunity. To date, two main classes of NS2B/NS3 of DENV and ZIKV protease inhibitors have been discovered: those that bind to the orthosteric site and those that act at the allosteric site. Therefore, this perspective article aims to discuss the main features of the use of the most potent NS2B/NS3 inhibitors and their impact at the social level.


Asunto(s)
Antivirales , Dengue , Inhibidores de Proteasas , Infección por el Virus Zika , Animales , Humanos , Antivirales/uso terapéutico , Antivirales/farmacología , ARN Helicasas DEAD-box , Dengue/tratamiento farmacológico , Dengue/virología , Virus del Dengue/efectos de los fármacos , Nucleósido-Trifosfatasa , Inhibidores de Proteasas/uso terapéutico , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteasas Virales , Virus Zika/efectos de los fármacos , Virus Zika/enzimología , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología
15.
J Cell Sci ; 137(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38660993

RESUMEN

Zika virus (ZIKV) has gained notoriety in recent years because there are no targeted therapies or vaccines available so far. Caveolin-1 (Cav-1) in host cells plays crucial functions in the invasion of many viruses. However, its specific involvement in ZIKV infection has remained unclear. Here, we reveal that depleting Cav-1 leads to a substantial reduction in ZIKV RNA levels, protein expression and viral particle production, indicating that ZIKV exploits Cav-1 for its infection. By dissecting each stage of the viral life cycle, we unveil that, unlike its invasion role in many other viruses, Cav-1 depletion selectively impairs ZIKV replication, resulting in altered replication dynamics and reduced strand-specific RNA levels, but does not affect viral entry, maturation and release. These results reveal an unforeseen function of Cav-1 in facilitating ZIKV replication, which provides new insights into the intricate interaction between Cav-1 and ZIKV and underscores Cav-1 as a potential candidate for anti-ZIKV approaches.


Asunto(s)
Caveolina 1 , ARN Viral , Replicación Viral , Infección por el Virus Zika , Virus Zika , Caveolina 1/metabolismo , Caveolina 1/genética , Virus Zika/fisiología , Virus Zika/metabolismo , Humanos , Infección por el Virus Zika/virología , Infección por el Virus Zika/metabolismo , ARN Viral/metabolismo , ARN Viral/genética , Animales , Interacciones Huésped-Patógeno , Chlorocebus aethiops , Células Vero , Células HEK293 , Internalización del Virus , Replicación de ARN
16.
Viruses ; 16(4)2024 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-38675903

RESUMEN

Gliomas account for approximately 75-80% of all malignant primary tumors in the central nervous system (CNS), with glioblastoma multiforme (GBM) considered the deadliest. Despite aggressive treatment involving a combination of chemotherapy, radiotherapy, and surgical intervention, patients with GBM have limited survival rates of 2 to 5 years, accompanied by a significant decline in their quality of life. In recent years, novel management strategies have emerged, such as immunotherapy, which includes the development of vaccines or T cells with chimeric antigen receptors, and oncolytic virotherapy (OVT), wherein wild type (WT) or genetically modified viruses are utilized to selectively lyse tumor cells. In vitro and in vivo studies have shown that the Zika virus (ZIKV) can infect glioma cells and induce a robust oncolytic activity. Consequently, interest in exploring this virus as a potential oncolytic virus (OV) for high-grade gliomas has surged. Given that ZIKV actively circulates in Colombia, evaluating its neurotropic and oncolytic capabilities holds considerable national and international importance, as it may emerge as an alternative for treating highly complex gliomas. Therefore, this literature review outlines the generalities of GBM, the factors determining ZIKV's specific tropism for nervous tissue, and its oncolytic capacity. Additionally, we briefly present the progress in preclinical studies supporting the use of ZIKV as an OVT for gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Viroterapia Oncolítica , Virus Oncolíticos , Infección por el Virus Zika , Virus Zika , Viroterapia Oncolítica/métodos , Humanos , Virus Zika/fisiología , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Glioma/terapia , Glioma/virología , Animales , Infección por el Virus Zika/terapia , Infección por el Virus Zika/virología , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virología , Glioblastoma/terapia , Glioblastoma/virología
17.
Viruses ; 16(4)2024 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-38675911

RESUMEN

Zika virus (ZIKV) remains a public health concern, with epidemics in endemic regions and sporadic outbreaks in new areas posing significant threats. Several mosquito-borne flaviviruses that can cause human illness, including West Nile, Usutu, and St. Louis encephalitis, have associations with birds. However, the susceptibility of chickens to ZIKV and their role in viral epidemiology is not currently known. We investigated the susceptibility of chickens to experimental ZIKV infection using chickens ranging from 1-day-old chicks to 6-week-old birds. ZIKV caused no clinical signs in chickens of all age groups tested. Viral RNA was detected in the blood and tissues during the first 5 days post-inoculation in 1-day and 4-day-old chicks inoculated with a high viral dose, but ZIKV was undetectable in 6-week-old birds at all timepoints. Minimal antibody responses were observed in 6-week-old birds, and while present in younger chicks, they waned by 28 days post-infection. Innate immune responses varied significantly between age groups. Robust type I interferon and inflammasome responses were measured in older chickens, while limited innate immune activation was observed in younger chicks. Signal transducer and activator of transcription 2 (STAT2) is a major driver of host restriction to ZIKV, and chicken STAT2 is distinct from human STAT2, potentially contributing to the observed resistance to ZIKV infection. The rapid clearance of the virus in older chickens coincided with an effective innate immune response, highlighting age-dependent susceptibility. Our study indicates that chickens are not susceptible to productive ZIKV infection and are unlikely to play a role in the ZIKV epidemiology.


Asunto(s)
Pollos , Inmunidad Innata , Enfermedades de las Aves de Corral , Infección por el Virus Zika , Virus Zika , Animales , Pollos/virología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Virus Zika/inmunología , Susceptibilidad a Enfermedades , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/inmunología , Factores de Edad , Anticuerpos Antivirales/sangre , ARN Viral/genética
18.
Viruses ; 16(4)2024 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-38675970

RESUMEN

Treating brain disease is challenging, and the Zika virus (ZIKV) presents a unique obstacle due to its neuroinvasive nature. In this review, we discuss the immunopathogenesis of ZIKV and explore how the virus interacts with the body's immune responses and the role of the protein Mfsd2a in maintaining the integrity of the blood-brain barrier (BBB) during ZIKV neuroinvasion. ZIKV has emerged as a significant public health concern due to its association with severe neurological problems, including microcephaly and Gillain-Barré Syndrome (GBS). Understanding its journey through the brain-particularly its interaction with the placenta and BBB-is crucial. The placenta, which is designed to protect the fetus, becomes a pathway for ZIKV when infected. The BBB is composed of brain endothelial cells, acts as a second barrier, and protects the fetal brain. However, ZIKV finds ways to disrupt these barriers, leading to potential damage. This study explores the mechanisms by which ZIKV enters the CNS and highlights the role of transcytosis, which allows the virus to move through the cells without significantly disrupting the BBB. Although the exact mechanisms of transcytosis are unclear, research suggests that ZIKV may utilize this pathway.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales , Transcitosis , Infección por el Virus Zika , Virus Zika , Humanos , Virus Zika/fisiología , Barrera Hematoencefálica/virología , Barrera Hematoencefálica/metabolismo , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología , Células Endoteliales/virología , Células Endoteliales/metabolismo , Animales , Embarazo , Femenino , Permeabilidad Capilar , Placenta/virología , Placenta/metabolismo , Encéfalo/virología , Encéfalo/metabolismo
19.
Antiviral Res ; 225: 105855, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38460762

RESUMEN

Zika virus (ZIKV) is an enveloped, single-stranded and positive-stranded RNA virus of the genus Flavivirus in the family Flaviviridae. ZIKV can cross the placental barrier and infect the fetus, causing microcephaly, congenital ZIKV syndrome, and even fetal death. ZIKV infection can also lead to testicular damage and male sterility. But no effective drugs and vaccines are available up to now. Previous studies have shown that the cathelicidin antimicrobial peptide LL-37 can protect against ZIKV infection. However, LL-37 is a secreted peptide, which can be easily degraded in vivo. We herein constructed exosome-loaded LL-37 (named LL-37-TM-exo and TM-LL-37-exo) using the transmembrane protein TM to load LL-37 onto the membrane of exosome. We found that exosome-loaded LL-37 could significantly inhibit ZIKV infection in vitro and in vivo, and LL-37-TM-exo had stronger antiviral activity than that of TM-LL-37-exo, which could significantly reduce ZIKV-induced testicular injury and sperm injury, and had broad-spectrum antiviral effect. Compared to free LL-37, exosome-loaded LL-37 showed a better serum stability, higher efficiency to cross the placental barrier, and stronger antiviral activity. The mechanism of exosome-loaded LL-37 against ZIKV infection was consistent with that of free LL-37, which could directly inactivate viral particles, reduce the susceptibility of host cells, and act on viral replication stage. Our study provides a novel strategy for the development of LL-37 against viral infection.


Asunto(s)
Catelicidinas , Exosomas , Infección por el Virus Zika , Virus Zika , Femenino , Humanos , Masculino , Embarazo , Antivirales/uso terapéutico , Exosomas/metabolismo , Placenta , Replicación Viral , Virus Zika/efectos de los fármacos , Virus Zika/fisiología , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/virología
20.
EMBO J ; 43(9): 1690-1721, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38378891

RESUMEN

Mosquitoes transmit many disease-relevant flaviviruses. Efficient viral transmission to mammalian hosts requires mosquito salivary factors. However, the specific salivary components facilitating viral transmission and their mechanisms of action remain largely unknown. Here, we show that a female mosquito salivary gland-specific protein, here named A. aegypti Neutrophil Recruitment Protein (AaNRP), facilitates the transmission of Zika and dengue viruses. AaNRP promotes a rapid influx of neutrophils, followed by virus-susceptible myeloid cells toward mosquito bite sites, which facilitates establishment of local infection and systemic dissemination. Mechanistically, AaNRP engages TLR1 and TLR4 of skin-resident macrophages and activates MyD88-dependent NF-κB signaling to induce the expression of neutrophil chemoattractants. Inhibition of MyD88-NF-κB signaling with the dietary phytochemical resveratrol reduces AaNRP-mediated enhancement of flavivirus transmission by mosquitoes. These findings exemplify how salivary components can aid viral transmission, and suggest a potential prophylactic target.


Asunto(s)
Aedes , Virus Zika , Animales , Aedes/virología , Aedes/metabolismo , Femenino , Virus Zika/fisiología , Ratones , Virus del Dengue/fisiología , Proteínas y Péptidos Salivales/metabolismo , Mosquitos Vectores/virología , Proteínas de Insectos/metabolismo , Células Mieloides/virología , Células Mieloides/metabolismo , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología , Infección por el Virus Zika/metabolismo , Dengue/transmisión , Dengue/virología , Dengue/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA