Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.044
Filtrar
1.
Parasit Vectors ; 17(1): 267, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918848

RESUMO

BACKGROUND: Past findings demonstrate that arthropods can egest midgut microbiota into the host skin leading to dual colonization of the vertebrate host with pathogens and saliva microbiome. A knowledge gap exists on how the saliva microbiome interacts with the pathogen in the saliva. To fill this gap, we need to first define the microbial composition of mosquito saliva. METHODS: The current study aimed at analyzing and comparing the microbial profile of Aedes albopictus saliva and midgut as well as assessing the impact of Zika virus (ZIKV) infection on the midgut and saliva microbial composition. Colony-reared Ae. albopictus strains were either exposed to ZIKV infectious or noninfectious bloodmeal. At 14 ays postinfection, the 16S V3-V4 hypervariable rRNA region was amplified from midgut and saliva samples and sequenced on an Illumina MiSeq platform. The relative abundance and diversity of midgut and saliva microbial taxa were assessed. RESULTS: We observed a richer microbial community in the saliva compared with the midgut, yet some of the microbial taxa were common in the midgut and saliva. ZIKV infection did not impact the microbial diversity of midgut or saliva. Further, we identified Elizabethkingia spp. in the Ae. albopictus saliva. CONCLUSIONS: This study provides insights into the microbial community of the Ae. albopictus saliva as well as the influence of ZIKV infection on the microbial composition of its midgut and saliva. The identification of Elizabethkingia spp., an emerging pathogen of global health significance, in Ae. albopictus saliva is of medical importance. Future studies to assess the interactions between Ae. albopictus saliva microbiome and ZIKV could lead to novel strategies for developing transmission barrier tools.


Assuntos
Aedes , Microbiota , Mosquitos Vetores , Saliva , Zika virus , Animais , Saliva/microbiologia , Saliva/virologia , Aedes/microbiologia , Aedes/virologia , Zika virus/genética , Zika virus/isolamento & purificação , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , Microbioma Gastrointestinal , RNA Ribossômico 16S/genética , Feminino , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/virologia
2.
Mar Drugs ; 22(6)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38921558

RESUMO

Considering the lack of antiviral drugs worldwide, we investigated the antiviral potential of fucoxanthin, an edible carotenoid purified from Sargassum siliquastrum, against zika virus (ZIKV) infection. The antiviral activity of fucoxanthin was assessed in ZIKV-infected Vero E6 cells, and the relevant structural characteristics were confirmed using molecular docking and molecular dynamics (MD) simulation. Fucoxanthin decreased the infectious viral particles and nonstructural protein (NS)1 mRNA expression levels at concentrations of 12.5, 25, and 50 µM in ZIKV-infected cells. Fucoxanthin also decreased the increased mRNA levels of interferon-induced proteins with tetratricopeptide repeat 1 and 2 in ZIKV-infected cells. Molecular docking simulations revealed that fucoxanthin binds to three main ZIKV proteins, including the envelope protein, NS3, and RNA-dependent RNA polymerase (RdRp), with binding energies of -151.449, -303.478, and -290.919 kcal/mol, respectively. The complex of fucoxanthin with RdRp was more stable than RdRp protein alone based on MD simulation. Further, fucoxanthin bonded to the three proteins via repeated formation and disappearance of hydrogen bonds. Overall, fucoxanthin exerts antiviral potential against ZIKV by affecting its three main proteins in a concentration-dependent manner. Thus, fucoxanthin isolated from S. siliquastrum is a potential candidate for treating zika virus infections.


Assuntos
Antivirais , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Sargassum , Xantofilas , Zika virus , Antivirais/farmacologia , Antivirais/isolamento & purificação , Antivirais/química , Zika virus/efeitos dos fármacos , Animais , Sargassum/química , Chlorocebus aethiops , Xantofilas/farmacologia , Xantofilas/isolamento & purificação , Xantofilas/química , Células Vero , Infecção por Zika virus/tratamento farmacológico , Infecção por Zika virus/virologia
3.
Nat Commun ; 15(1): 5426, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926343

RESUMO

Zika and dengue virus nonstructural protein 5 antagonism of STAT2, a critical interferon signaling transcription factor, to suppress the host interferon response is required for viremia and pathogenesis in a vertebrate host. This affects viral species tropism, as mouse STAT2 resistance renders only immunocompromised or humanized STAT2 mice infectable. Here, we explore how STAT2 evolution impacts antagonism. By measuring the susceptibility of 38 diverse STAT2 proteins, we demonstrate that resistance arose numerous times in mammalian evolution. In four species, resistance requires distinct sets of multiple amino acid changes that often individually disrupt STAT2 signaling. This reflects an evolutionary ridge where progressive resistance is balanced by the need to maintain STAT2 function. Furthermore, resistance may come with a fitness cost, as resistance that arose early in lemur evolution was subsequently lost in some lemur lineages. These findings underscore that while it is possible to evolve resistance to antagonism, complex evolutionary trajectories are required to avoid detrimental host fitness consequences.


Assuntos
Evolução Molecular , Fator de Transcrição STAT2 , Proteínas não Estruturais Virais , Fator de Transcrição STAT2/metabolismo , Fator de Transcrição STAT2/genética , Animais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Humanos , Camundongos , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Zika virus/genética , Flavivirus/genética , Flavivirus/fisiologia , Filogenia , Interações Hospedeiro-Patógeno/genética
4.
Viruses ; 16(6)2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38932129

RESUMO

The complete lack of yellow fever virus (YFV) in Asia, and the lack of urban YFV transmission in South America, despite the abundance of the peridomestic mosquito vector Aedes (Stegomyia.) aegypti is an enigma. An immunologically naïve population of over 2 billion resides in Asia, with most regions infested with the urban YF vector. One hypothesis for the lack of Asian YF, and absence of urban YF in the Americas for over 80 years, is that prior immunity to related flaviviruses like dengue (DENV) or Zika virus (ZIKV) modulates YFV infection and transmission dynamics. Here we utilized an interferon α/ß receptor knock-out mouse model to determine the role of pre-existing dengue-2 (DENV-2) and Zika virus (ZIKV) immunity in YF virus infection, and to determine mechanisms of cross-protection. We utilized African and Brazilian YF strains and found that DENV-2 and ZIKV immunity significantly suppresses YFV viremia in mice, but may or may not protect relative to disease outcomes. Cross-protection appears to be mediated mainly by humoral immune responses. These studies underscore the importance of re-assessing the risks associated with YF outbreak while accounting for prior immunity from flaviviruses that are endemic.


Assuntos
Proteção Cruzada , Vírus da Dengue , Modelos Animais de Doenças , Camundongos Knockout , Receptor de Interferon alfa e beta , Febre Amarela , Vírus da Febre Amarela , Infecção por Zika virus , Zika virus , Animais , Febre Amarela/imunologia , Febre Amarela/prevenção & controle , Febre Amarela/virologia , Camundongos , Proteção Cruzada/imunologia , Vírus da Febre Amarela/imunologia , Zika virus/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/virologia , Vírus da Dengue/imunologia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/deficiência , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Flavivirus/imunologia , Aedes/virologia , Aedes/imunologia , Dengue/imunologia , Dengue/prevenção & controle , Dengue/virologia , Feminino , Viremia/imunologia , Mosquitos Vetores/virologia , Mosquitos Vetores/imunologia , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/prevenção & controle , Infecções por Flavivirus/virologia , Camundongos Endogâmicos C57BL
5.
Viruses ; 16(6)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38932158

RESUMO

Humans continue to be at risk from the Zika virus. Although there have been significant research advancements regarding Zika, the absence of a vaccine or approved treatment poses further challenges for healthcare providers. In this study, we developed a microparticulate Zika vaccine using an inactivated whole Zika virus as the antigen that can be administered pain-free via intranasal (IN) immunization. These microparticles (MP) were formulated using a double emulsion method developed by our lab. We explored a prime dose and two-booster-dose vaccination strategy using MPL-A® and Alhydrogel® as adjuvants to further stimulate the immune response. MPL-A® induces a Th1-mediated immune response and Alhydrogel® (alum) induces a Th2-mediated immune response. There was a high recovery yield of MPs, less than 5 µm in size, and particle charge of -19.42 ± 0.66 mV. IN immunization of Zika MP vaccine and the adjuvanted Zika MP vaccine showed a robust humoral response as indicated by several antibodies (IgA, IgM, and IgG) and several IgG subtypes (IgG1, IgG2a, and IgG3). Vaccine MP elicited a balance Th1- and Th2-mediated immune response. Immune organs, such as the spleen and lymph nodes, exhibited a significant increase in CD4+ helper and CD8+ cytotoxic T-cell cellular response in both vaccine groups. Zika MP vaccine and adjuvanted Zika MP vaccine displayed a robust memory response (CD27 and CD45R) in the spleen and lymph nodes. Adjuvanted vaccine-induced higher Zika-specific intracellular cytokines than the unadjuvanted vaccine. Our results suggest that more than one dose or multiple doses may be necessary to achieve necessary immunological responses. Compared to unvaccinated mice, the Zika vaccine MP and adjuvanted MP vaccine when administered via intranasal route demonstrated robust humoral, cellular, and memory responses. In this pre-clinical study, we established a pain-free microparticulate Zika vaccine that produced a significant immune response when administered intranasally.


Assuntos
Administração Intranasal , Anticorpos Antivirais , Vacinas Virais , Infecção por Zika virus , Zika virus , Animais , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/imunologia , Zika virus/imunologia , Camundongos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Feminino , Imunização/métodos , Adjuvantes Imunológicos/administração & dosagem , Modelos Animais de Doenças , Adjuvantes de Vacinas/administração & dosagem , Vacinação/métodos , Citocinas/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia
6.
Molecules ; 29(11)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38893293

RESUMO

Within the fields of infectious disease diagnostics, microfluidic-based integrated technology systems have become a vital technology in enhancing the rapidity, accuracy, and portability of pathogen detection. These systems synergize microfluidic techniques with advanced molecular biology methods, including reverse transcription polymerase chain reaction (RT-PCR), loop-mediated isothermal amplification (LAMP), and clustered regularly interspaced short palindromic repeats (CRISPR), have been successfully used to identify a diverse array of pathogens, including COVID-19, Ebola, Zika, and dengue fever. This review outlines the advances in pathogen detection, attributing them to the integration of microfluidic technology with traditional molecular biology methods and smartphone- and paper-based diagnostic assays. The cutting-edge diagnostic technologies are of critical importance for disease prevention and epidemic surveillance. Looking ahead, research is expected to focus on increasing detection sensitivity, streamlining testing processes, reducing costs, and enhancing the capability for remote data sharing. These improvements aim to achieve broader coverage and quicker response mechanisms, thereby constructing a more robust defense for global public health security.


Assuntos
Técnicas de Diagnóstico Molecular , Técnicas de Amplificação de Ácido Nucleico , Humanos , Técnicas de Amplificação de Ácido Nucleico/métodos , Técnicas de Diagnóstico Molecular/métodos , Microfluídica/métodos , Doenças Transmissíveis/diagnóstico , COVID-19/diagnóstico , COVID-19/virologia , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , Técnicas Analíticas Microfluídicas/métodos , Dengue/diagnóstico , Infecção por Zika virus/diagnóstico , Infecção por Zika virus/virologia , Zika virus/genética , Zika virus/isolamento & purificação
7.
Gigascience ; 132024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38869150

RESUMO

Viral helicases are promising targets for the development of antiviral therapies. Given their vital function of unwinding double-stranded nucleic acids, inhibiting them blocks the viral replication cycle. Previous studies have elucidated key structural details of these helicases, including the location of substrate binding sites, flexible domains, and the discovery of potential inhibitors. Here we present a series of new Galaxy tools and workflows for performing and analyzing molecular dynamics simulations of viral helicases. We first validate them by demonstrating recapitulation of data from previous simulations of Zika (NS3) and SARS-CoV-2 (NSP13) helicases in apo and complex with inhibitors. We further demonstrate the utility and generalizability of these Galaxy workflows by applying them to new cases, proving their usefulness as a widely accessible method for exploring antiviral activity.


Assuntos
Simulação de Dinâmica Molecular , SARS-CoV-2 , SARS-CoV-2/enzimologia , Zika virus/enzimologia , Fluxo de Trabalho , RNA Helicases/química , RNA Helicases/metabolismo , Humanos , DNA Helicases/química , DNA Helicases/metabolismo , Antivirais/química , Antivirais/farmacologia , Proteases Semelhantes à Papaína de Coronavírus/química , Proteases Semelhantes à Papaína de Coronavírus/metabolismo , Sítios de Ligação , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo
8.
Lancet Glob Health ; 12(7): e1129-e1138, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38876760

RESUMO

BACKGROUND: Data on long-term neurodevelopmental outcomes of normocephalic children (born with normal head circumference) exposed to Zika virus in utero are scarce. We aimed to compare neurodevelopmental outcomes in normocephalic children up to age 48 months with and without Zika virus exposure in utero. METHODS: In this prospective cohort study, we included infants from two cohorts of normocephalic children born in León and Managua, Nicaragua during the 2016 Zika epidemic. In León, all women pregnant during the two enrolment periods were eligible. In Managua, mother-child pairs were included from three districts in the municipality of Managua: all women who became pregnant before June 15, 2016, and had a due date of Sept 15, 2016 or later were eligible. Infants were serologically classified as Zika virus-exposed or Zika virus-unexposed in utero and were followed up prospectively until age 48 months. At 36 months and 48 months of age, the Mullen Scales of Early Learning (MSEL) assessment was administered. Primary outcomes were MSEL early learning composite (ELC) scores at 30-48 months in León and 36-48 months in Managua. We used an inverse probability weighting generalised estimating equations model to assess the effect of Zika virus exposure on individual MSEL cognitive domain scores and ELC scores, adjusted for maternal education and age, poverty status, and infant sex. FINDINGS: The initial enrolment period for the León cohort was between Jan 31 and April 5, 2017 and the second was between Aug 30, 2017, and Feb 22, 2018. The enrolment period for the Managua cohort was between Oct 24, 2019, and May 5, 2020. 478 mothers (482 infants) from the León cohort and 615 mothers (609 infants) from the Managua cohort were enrolled, of whom 622 children (303 from the León cohort; 319 from the Managua cohort) were included in the final analysis; four children had microcephaly at birth and thus were excluded from analyses, two from each cohort. 33 (11%) of 303 children enrolled in León and 219 (69%) of 319 children enrolled in Managua were exposed to Zika virus in utero. In both cohorts, no significant differences were identified in adjusted mean ELC scores between Zika virus-exposed and unexposed infants at 36 months (between-group difference 1·2 points [95% CI -4·2 to 6·5] in the León cohort; 2·8 [-2·4 to 8·1] in the Managua cohort) or at 48 months (-0·9 [-10·8 to 8·8] in the León cohort; 0·1 [-5·1 to 5·2] in the Managua cohort). No differences in ELC scores between Zika virus-exposed and unexposed infants exceeded 6 points at any time between 30 months and 48 months in León or between 36 months and 48 months in Managua, which was considered clinically significant in other settings. INTERPRETATION: We found no significant differences in neurodevelopmental scores between normocephalic children with in-utero Zika virus exposure and Zika virus-unexposed children at age 36 months or 48 months. These findings are promising, supporting typical neurodevelopment in Zika virus-exposed normocephalic children, although additional follow-up and research is warranted. FUNDING: National Institute of Child Health and Development, National Institute of Allergy and Infectious Diseases, and Fogarty International Center. TRANSLATION: For the Spanish translation of the abstract see Supplementary Materials section.


Assuntos
Desenvolvimento Infantil , Complicações Infecciosas na Gravidez , Efeitos Tardios da Exposição Pré-Natal , Infecção por Zika virus , Humanos , Nicarágua/epidemiologia , Infecção por Zika virus/epidemiologia , Feminino , Estudos Prospectivos , Pré-Escolar , Gravidez , Masculino , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Efeitos Tardios da Exposição Pré-Natal/virologia , Lactente , Complicações Infecciosas na Gravidez/epidemiologia , Complicações Infecciosas na Gravidez/virologia , Zika virus , Adulto , Transtornos do Neurodesenvolvimento/epidemiologia , Transtornos do Neurodesenvolvimento/virologia
9.
Nat Commun ; 15(1): 5173, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890352

RESUMO

Zika virus (ZikV) infection during pregnancy can cause congenital Zika syndrome (CZS) and neurodevelopmental delay in infants, of which the pathogenesis remains poorly understood. We utilize an established female pigtail macaque maternal-to-fetal ZikV infection/exposure model to study fetal brain pathophysiology of CZS manifesting from ZikV exposure in utero. We find prenatal ZikV exposure leads to profound disruption of fetal myelin, with extensive downregulation in gene expression for key components of oligodendrocyte maturation and myelin production. Immunohistochemical analyses reveal marked decreases in myelin basic protein intensity and myelinated fiber density in ZikV-exposed animals. At the ultrastructural level, the myelin sheath in ZikV-exposed animals shows multi-focal decompaction, occurring concomitant with dysregulation of oligodendrocyte gene expression and maturation. These findings define fetal neuropathological profiles of ZikV-linked brain injury underlying CZS resulting from ZikV exposure in utero. Because myelin is critical for cortical development, ZikV-related perturbations in oligodendrocyte function may have long-term consequences on childhood neurodevelopment, even in the absence of overt microcephaly.


Assuntos
Modelos Animais de Doenças , Bainha de Mielina , Oligodendroglia , Infecção por Zika virus , Zika virus , Animais , Infecção por Zika virus/virologia , Infecção por Zika virus/patologia , Oligodendroglia/virologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Feminino , Bainha de Mielina/metabolismo , Gravidez , Zika virus/patogenicidade , Complicações Infecciosas na Gravidez/virologia , Complicações Infecciosas na Gravidez/patologia , Macaca nemestrina , Encéfalo/virologia , Encéfalo/patologia , Encéfalo/metabolismo , Humanos , Proteína Básica da Mielina/metabolismo , Proteína Básica da Mielina/genética
10.
Arch Virol ; 169(7): 135, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38839691

RESUMO

Monocytes are the primary targets of Zika virus (ZIKV) and are associated with ZIKV pathogenesis. Currently, there is no effective treatment for ZIKV infection. It is known that 1,25-dihydroxy vitamin D3 (VitD3) has strong antiviral activity in dengue virus-infected macrophages, but it is unknown whether VitD3 inhibits ZIKV infection in monocytes. We investigated the relationship between ZIKV infection and the expression of genes of the VitD3 pathway, as well as the inflammatory response of infected monocytes in vitro. ZIKV replication was evaluated using a plaque assay, and VitD3 pathway gene expression was analyzed by RT-qPCR. Pro-inflammatory cytokines/chemokines were quantified using ELISA. We found that VitD3 did not suppress ZIKV replication. The results showed a significant decrease in the expression of vitamin D3 receptor (VDR), cytochrome P450 family 24 subfamily A member 1 (CYP24A1), and cathelicidin antimicrobial peptide (CAMP) genes upon ZIKV infection. Treatment with VitD3 was unable to down-modulate production of pro-inflammatory cytokines, except TNF-α, and chemokines. This suggests that ZIKV infection inhibits the expression of VitD3 pathway genes, thereby preventing VitD3-dependent inhibition of viral replication and the inflammatory response. This is the first study to examine the effects of VitD3 in the context of ZIKV infection, and it has important implications for the role of VitD3 in the control of viral replication and inflammatory responses during monocyte infection.


Assuntos
Catelicidinas , Monócitos , Replicação Viral , Vitamina D3 24-Hidroxilase , Infecção por Zika virus , Zika virus , Humanos , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Citocinas/metabolismo , Citocinas/genética , Monócitos/virologia , Monócitos/metabolismo , Monócitos/imunologia , Receptores de Calcitriol/metabolismo , Receptores de Calcitriol/genética , Replicação Viral/efeitos dos fármacos , Vitamina D3 24-Hidroxilase/genética , Vitamina D3 24-Hidroxilase/metabolismo , Zika virus/fisiologia , Infecção por Zika virus/virologia , Infecção por Zika virus/metabolismo
11.
Sci Transl Med ; 16(749): eadn2199, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38809964

RESUMO

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.


Assuntos
Vírus da Dengue , Dengue , Sorogrupo , Infecção por Zika virus , Zika virus , Humanos , Zika virus/imunologia , Dengue/imunologia , Dengue/virologia , Vírus da Dengue/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Criança , Feminino , Masculino , Nicarágua/epidemiologia , Pré-Escolar , Fatores de Risco , Adolescente , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Estudos de Coortes
12.
Sci Signal ; 17(837): eadi9844, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38771918

RESUMO

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.


Assuntos
2',5'-Oligoadenilato Sintetase , Endorribonucleases , RNA de Cadeia Dupla , Zika virus , Endorribonucleases/metabolismo , Endorribonucleases/genética , Endorribonucleases/química , Humanos , 2',5'-Oligoadenilato Sintetase/metabolismo , 2',5'-Oligoadenilato Sintetase/genética , 2',5'-Oligoadenilato Sintetase/química , RNA de Cadeia Dupla/metabolismo , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/genética , Zika virus/metabolismo , Animais , Vírus da Dengue/metabolismo , RNA Viral/metabolismo , RNA Viral/genética , Estabilidade de RNA , Vírus do Nilo Ocidental/metabolismo , Vírus do Nilo Ocidental/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia , Ativação Enzimática , Células HeLa , Células HEK293
13.
Sci Rep ; 14(1): 12533, 2024 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822066

RESUMO

In flaviviruses such as Dengue or Zika, non-structural (NS) NS4A protein forms homo-oligomers, participates in membrane remodelling and is critical for virulence. In both viruses, mature NS4A has the same length and three predicted hydrophobic domains. The oligomers formed by Dengue NS4A are reported to be small (n = 2, 3), based on denaturing SDS gels, but no high-resolution structure of a flavivirus NS4A protein is available, and the size of the oligomer in lipid membranes is not known. Herein we show that crosslinking Zika NS4A protein in lipid membranes results in oligomers at least up to hexamers. Further, sedimentation velocity shows that NS4A in mild detergent C14-betaine appears to be in fast equilibrium between at least two species, where one is smaller, and the other larger, than a trimer or a tetramer. Consistently, sedimentation equilibrium data was best fitted to a model involving an equilibrium between dimers (n = 2) and hexamers (n = 6). Overall, the large, at least hexameric, oligomers obtained herein in liposomes and in mild detergent are more likely to represent the forms of NS4A present in cell membranes.


Assuntos
Detergentes , Lipossomos , Multimerização Proteica , Proteínas não Estruturais Virais , Zika virus , Lipossomos/química , Lipossomos/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Detergentes/química , Zika virus/química
14.
Vaccine ; 42(17): 3674-3683, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38749821

RESUMO

The Zika virus (ZIKV) is considered a public health problem worldwide due to its association with the development of microcephaly and the Guillain-Barré syndrome. Currently, there is no specific treatment or vaccine approved to combat this disease, and thus, developing safe and effective vaccines is a relevant goal. In this study, a multi-epitope protein called rpZDIII was designed based on a series of ZIKV antigenic sequences, a bacterial carrier, and linkers. The analysis of the predicted 3D structure of the rpZDIII chimeric antigen was performed on the AlphaFold 2 server, and it was produced in E. coli and purified from inclusion bodies, followed by solubilization and refolding processes. The yield achieved for rpZDIII was 11 mg/L in terms of pure soluble recombinant protein per liter of fermentation. rpZDIII was deemed immunogenic since it induced serum IgG and IgM responses in mice upon subcutaneous immunization in a three-dose scheme. Moreover, sera from mice immunized with rpZDIII showed neutralizing activity against ZIKV. Therefore, this study reveals rpZDIII as a promising immunogen for the development of a rationally designed multi-epitope vaccine against ZIKV, and completion of its preclinical evaluation is guaranteed.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Antígenos Virais , Infecção por Zika virus , Zika virus , Animais , Zika virus/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Camundongos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/imunologia , Antígenos Virais/imunologia , Antígenos Virais/genética , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Epitopos/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Feminino , Escherichia coli/genética , Escherichia coli/metabolismo , Imunoglobulina M/imunologia , Imunoglobulina M/sangue , Camundongos Endogâmicos BALB C
15.
J Virol ; 98(6): e0170523, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38742902

RESUMO

Long non-coding RNAs (lncRNAs) represent a new group of host factors involved in viral infection. Current study identified an intergenic lncRNA, LINC08148, as a proviral factor of Zika virus (ZIKV) and Dengue virus 2 (DENV2). Knockout (KO) or silencing of LINC08148 decreases the replication of ZIKV and DENV2. LINC08148 mainly acts at the endocytosis step of ZIKV but at a later stage of DENV2. RNA-seq analysis reveals that LINC08148 knockout downregulates the transcription levels of five endocytosis-related genes including AP2B1, CHMP4C, DNM1, FCHO1, and Src. Among them, loss of Src significantly decreases the uptake of ZIKV. Trans-complementation of Src in the LINC08148KO cells largely restores the caveola-mediated endocytosis of ZIKV, indicating that the proviral effect of LINC08148 is exerted through Src. Finally, LINC08148 upregulates the Src transcription through associating with its transcription factor SP1. This work establishes an essential role of LINC08148 in the ZIKV entry, underscoring a significance of lncRNAs in the viral infection. IMPORTANCE: Long non-coding RNAs (lncRNAs), like proteins, participate in viral infection. However, functions of most lncRNAs remain unknown. In this study, we performed a functional screen based on microarray data and identified a new proviral lncRNA, LINC08148. Then, we uncovered that LINC08148 is involved in the caveola-mediated endocytosis of ZIKV, rather than the classical clathrin-mediated endocytosis. Mechanistically, LINC08148 upregulates the transcription of Src, an initiator of caveola-mediated endocytosis, through binding to its transcription factor SP1. This study identifies a new lncRNA involved in the ZIKV infection, suggesting lncRNAs and cellular proteins are closely linked and cooperate to regulate viral infection.


Assuntos
Endocitose , RNA Longo não Codificante , Internalização do Vírus , Infecção por Zika virus , Zika virus , RNA Longo não Codificante/metabolismo , RNA Longo não Codificante/genética , Zika virus/genética , Zika virus/fisiologia , Humanos , Infecção por Zika virus/virologia , Infecção por Zika virus/metabolismo , Infecção por Zika virus/genética , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp1/genética , Cavéolas/metabolismo , Animais , Replicação Viral , Regulação para Cima , Vírus da Dengue/fisiologia , Vírus da Dengue/genética , Chlorocebus aethiops , Células HEK293 , Células Vero , Quinases da Família src/metabolismo , Quinases da Família src/genética
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(5): 447-454, 2024 May.
Artigo em Chinês | MEDLINE | ID: mdl-38790101

RESUMO

Objective To prepare monoclonal antibodies against the envelope protein extracellular domain (Eecto) of Zika virus (ZIKV) in mice. Methods A prokaryotic expression plasmid, pET28a-ZIKV-Eecto of ZIKV Eecto, was constructed, transformed into Escherichia coli BL21 and induced by isopropyl ß-D-thiogalactoside (IPTG). The recombinant Eecto protein was expressed in the form of inclusion bodies, and purified proteins were obtained through denaturation, renaturation and ultrafiltration. After three rounds of immunization with the Eecto protein, the serum of BALB/c mice was obtained and the titer of polyclonal antibodies in serum was determined. The reactivity of polyclonal antibodies was analyzed with Western blotting and immunofluorescence assay in HEK293T cells expressing the ZIKV prME. Spleen cells from mice with higher antibody titers were prepared and fused with SP2/0 myeloma cells. The hybridoma cells secreting antibodies were screened through the limited dilution method, and the ascites containing antibody were harvested for titer measurement and subclass analysis. The Eecto from the envelope proteins of Japanese encephalitis virus (JEV), Yellow fever virus (YFV), Dengue virus (DENV1-4), and Tick borne encephalitis virus (TBEV) were coated and used to analyze the cross-reactivity of ZIKV monoclonal antibodies by ELISA. Further specificity analysis was conducted on antibodies with high titers and strong specificity. Results The plasmid pET28a-ZIKV-Eecto was successfully constructed. The purified Eecto protein was obtained with good immunogenicity. Four monoclonal antibodies were prepared and screened, namely 1D6, 4F11, 4H7, and 4F8. Among them, 1D6, 4H7, and 4F8 are IgG (K) type antibodies, and 4F11 is an IgM (K) antibody. The ascitic fluid titer of 1D6 was higher than 1:108. Antibodies 1D6 and 4H7 are ZIKV-specific and showed no cross-reactivity with other Flaviviruses. Conclusion The mice monoclonal antibodies against ZIKV-Eecto are produced successfully, which will provide experimental materials for the establishment of ZIKV detection methods and the study of its pathogenesis.


Assuntos
Anticorpos Monoclonais , Camundongos Endogâmicos BALB C , Proteínas do Envelope Viral , Zika virus , Animais , Zika virus/imunologia , Zika virus/genética , Anticorpos Monoclonais/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/genética , Camundongos , Humanos , Células HEK293 , Feminino , Anticorpos Antivirais/imunologia , Domínios Proteicos/imunologia , Ensaio de Imunoadsorção Enzimática
17.
Antiviral Res ; 227: 105915, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38777094

RESUMO

The genus of flavivirus includes many mosquito-borne human pathogens, such as Zika (ZIKV) and the four serotypes of dengue (DENV1-4) viruses, that affect billions of people as evidenced by epidemics and endemicity in many countries and regions in the world. Among the 10 viral proteins encoded by the viral genome, the nonstructural protein 1 (NS1) is the only secreted protein and has been used as a diagnostic biomarker. NS1 has also been an attractive target for its biotherapeutic potential as a vaccine antigen. This review focuses on the recent advances in the structural landscape of the secreted NS1 (sNS1) and its complex with monoclonal antibodies (mAbs). NS1 forms an obligatory dimer, and upon secretion, it has been reported to be hexametric (trimeric dimers) that could dissociate and bind to the epithelial cell membrane. However, high-resolution structural information has been missing about the high-order oligomeric states of sNS1. Several cryoEM studies have since shown that DENV and ZIKV recombinant sNS1 (rsNS1) are in dynamic equilibrium of dimer-tetramer-hexamer states, with tetramer being the predominant form. It was recently revealed that infection-derived sNS1 (isNS1) forms a complex of the NS1 dimer partially embedded in a High-Density Lipoprotein (HDL) particle. Structures of NS1 in complexes with mAbs have also been reported which shed light on their protective roles during infection. The biological significance of the diversity of NS1 oligomeric states remains to be further studied, to inform future research on flaviviral pathogenesis and the development of therapeutics and vaccines. Given the polymorphism of flavivirus NS1 across sample types with variations in antigenicity, we propose a nomenclature to accurately define NS1 based on the localization and origin.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais , Flavivirus , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Humanos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/química , Anticorpos Antivirais/imunologia , Flavivirus/imunologia , Flavivirus/química , Flavivirus/genética , Animais , Zika virus/imunologia , Zika virus/genética , Zika virus/química , Vírus da Dengue/imunologia , Vírus da Dengue/genética , Vírus da Dengue/química , Multimerização Proteica , Conformação Proteica
18.
ACS Appl Bio Mater ; 7(5): 2862-2871, 2024 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-38699864

RESUMO

Mosquito-borne viruses are a major worldwide health problem associated with high morbidity and mortality rates and significant impacts on national healthcare budgets. The development of antiviral drugs for both the treatment and prophylaxis of these diseases is thus of considerable importance. To address the need for therapeutics with antiviral activity, a library of heparan sulfate mimetic polymers was screened against dengue virus (DENV), Yellow fever virus (YFV), Zika virus (ZIKV), and Ross River virus (RRV). The polymers were prepared by RAFT polymerization of various acidic monomers with a target MW of 20 kDa (average Mn ∼ 27 kDa by GPC). Among the polymers, poly(SS), a homopolymer of sodium styrenesulfonate, was identified as a broad spectrum antiviral with activity against all the tested viruses and particularly potent inhibition of YFV (IC50 = 310 pM). Our results further uncovered that poly(SS) exhibited a robust inhibition of ZIKV infection in both mosquito and human cell lines, which points out the potential functions of poly(SS) in preventing mosquito-borne viruses associated diseases by blocking viral transmission in their mosquito vectors and mitigating viral infection in patients.


Assuntos
Antivirais , Heparitina Sulfato , Polímeros , Antivirais/farmacologia , Antivirais/química , Antivirais/síntese química , Heparitina Sulfato/química , Heparitina Sulfato/farmacologia , Animais , Humanos , Polímeros/química , Polímeros/farmacologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/síntese química , Culicidae/efeitos dos fármacos , Culicidae/virologia , Testes de Sensibilidade Microbiana , Teste de Materiais , Tamanho da Partícula , Linhagem Celular , Estrutura Molecular , Chlorocebus aethiops , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Zika virus/efeitos dos fármacos
19.
Talanta ; 276: 126215, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38723474

RESUMO

Antibody detection is the critical first step for tracking the spread of many diseases including COVID-19. Lateral flow immunoassay (LFIA) is the most commonly used method for rapid antibody detection because it is easy-to-use and inexpensive. However, LFIA has limited sensitivity when gold nanoparticles (AuNPs) are used as the signals. In this study, the endospores of Bacillus subtilis were used in combination with AuNP in a LFIA to detect antibodies. The endospores serve as a signal amplifier. The detection limit was about 10-8 M for anti-beta galactosidase antibody detection whereas the detection limit of conventional LFIA is about 10-6 M. Furthermore, the proposed methods have no additional user steps compared with the traditional LFIA. This method, therefore, improved the sensitivity 100-fold without compromising any advantages of LFIA. We believe that the proposed method will be useful for detection of antibodies against HIV, Zika virus, SARS-CoV-2, and so on.


Assuntos
Bacillus subtilis , Ouro , Limite de Detecção , Nanopartículas Metálicas , Bacillus subtilis/imunologia , Imunoensaio/métodos , Ouro/química , Nanopartículas Metálicas/química , Humanos , Esporos Bacterianos/imunologia , SARS-CoV-2/imunologia , Anticorpos Antivirais/imunologia , COVID-19/diagnóstico , COVID-19/imunologia , COVID-19/virologia , Zika virus/imunologia
20.
Anal Chem ; 96(22): 9244-9253, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38773697

RESUMO

Sensitive identification and effective inactivation of the virus are paramount for the early diagnosis and treatment of viral infections to prevent the risk of secondary transmission of viruses in the environment. Herein, we developed a novel two-step fluorescence immunoassay using antibody/streptavidin dual-labeled polystyrene nanobeads and biotin-labeled G-quadruplex/hemin DNAzymes with peroxidase-mimicking activity for sensitive quantitation and efficient inactivation of living Zika virus (ZIKV). The dual-labeled nanobeads can specifically bind ZIKV through E protein targeting and simultaneously accumulate DNAzymes, leading to the catalytic oxidation of Amplex Red indicators and generation of intensified aggregation-induced emission fluorescence signals, with a detection limit down to 66.3 PFU/mL and 100% accuracy. Furthermore, robust reactive oxygen species generated in situ by oxidized Amplex Red upon irradiation can completely kill the virus. This sensitive and efficient detection-inactivation integrated system will expand the viral diagnostic tools and reduce the risk of virus transmission in the environment.


Assuntos
DNA Catalítico , Zika virus , DNA Catalítico/química , DNA Catalítico/metabolismo , Imunoensaio/métodos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Limite de Detecção , Quadruplex G , Inativação de Vírus/efeitos da radiação , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...