Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Gen Virol ; 105(1)2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38180473

RESUMO

Nipah virus (NiV) is a deadly zoonotic pathogen with high potential to cause another pandemic. Owing to biosafety concerns, studies on living NiV must be performed in biosafety level 4 (BSL-4) laboratories, which greatly hinders the development of anti-NiV drugs. To overcome this issue, minigenome systems have been developed to study viral replication and screen for antiviral drugs. This study aimed to develop two minigenome systems (transient and stable expression) based on a helper cell line expressing the NiV P, N and L proteins required to initiate NiV RNA replication. Stable minigenome cells were resistant to ribavirin, remdesivir and favipiravir but sensitive to interferons. Cells of the transient replication system were sensitive to ribavirin and favipiravir and suitable for drug screening. Our study demonstrates a feasible and effective platform for studying NiV replication and shows great potential for high-throughput drug screening in a BSL-2 laboratory environment.


Assuntos
Vírus Nipah , Vírus Nipah/genética , Ribavirina , Replicação Viral , Antivirais/farmacologia
2.
Virol Sin ; 39(1): 56-70, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37967718

RESUMO

Avian H9N2 viruses have wide host range among the influenza A viruses. However, knowledge of H9N2 mammalian adaptation is limited. To explore the molecular basis of the adaptation to mammals, we performed serial lung passaging of the H9N2 strain A/chicken/Hunan/8.27 YYGK3W3-OC/2018 (3W3) in mice and identified six mutations in the hemagglutinin (HA) and polymerase acidic (PA) proteins. Mutations L226Q, T511I, and A528V of HA were responsible for enhanced pathogenicity and viral replication in mice; notably, HA-L226Q was the key determinant. Mutations T97I, I545V, and S594G of PA contributed to enhanced polymerase activity in mammalian cells and increased viral replication levels in vitro and in vivo. PA-T97I increased viral polymerase activity by accelerating the viral polymerase complex assembly. Our findings revealed that the viral replication was affected by the presence of PA-97I and/or PA-545V in combination with a triple-point HA mutation. Furthermore, the double- and triple-point PA mutations demonstrated antagonistic effect on viral replication when combined with HA-226Q. Notably, any combination of PA mutations, along with double-point HA mutations, resulted in antagonistic effect on viral replication. We also observed antagonism in viral replication between PA-545V and PA-97I, as well as between HA-528V and PA-545V. Our findings demonstrated that several antagonistic mutations in HA and PA proteins affect viral replication, which may contribute to the H9N2 virus adaptation to mice and mammalian cells. These findings can potentially contribute to the monitoring of H9N2 field strains for assessing their potential risk in mammals.


Assuntos
Vírus da Influenza A Subtipo H9N2 , Influenza Aviária , Infecções por Orthomyxoviridae , Animais , Camundongos , Vírus da Influenza A Subtipo H9N2/genética , Hemaglutininas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Mutação , Replicação Viral/genética , Nucleotidiltransferases , Galinhas , Mamíferos/metabolismo
3.
PLoS Pathog ; 19(9): e1011620, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37656756

RESUMO

LGP2 is a RIG-I-like receptor (RLR) known to bind and recognize the intermediate double-stranded RNA (dsRNA) during virus infection and to induce type-I interferon (IFN)-related antiviral innate immune responses. Here, we find that LGP2 inhibits Zika virus (ZIKV) and tick-borne encephalitis virus (TBEV) replication independent of IFN induction. Co-immunoprecipitation (Co-IP) and confocal immunofluorescence data suggest that LGP2 likely colocalizes with the replication complex (RC) of ZIKV by interacting with viral RNA-dependent RNA polymerase (RdRP) NS5. We further verify that the regulatory domain (RD) of LGP2 directly interacts with RdRP of NS5 by biolayer interferometry assay. Data from in vitro RdRP assays indicate that LGP2 may inhibit polymerase activities of NS5 at pre-elongation but not elongation stages, while an RNA-binding-defective LGP2 mutant can still inhibit RdRP activities and virus replication. Taken together, our work suggests that LGP2 can inhibit flavivirus replication through direct interaction with NS5 protein and downregulates its polymerase pre-elongation activities, demonstrating a distinct role of LGP2 beyond its function in innate immune responses.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Infecção por Zika virus , Zika virus , Humanos , RNA Polimerase Dependente de RNA/genética , Nucleotidiltransferases , RNA de Cadeia Dupla
5.
Food Environ Virol ; 15(2): 123-130, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36890342

RESUMO

SARS-CoV-2 contaminated items in the cold chain becomes a threat to public health, therefore the effective and safe sterilization method fit for the low temperature is needed. Ultraviolet is an effective sterilization method while its effect on SARS-CoV-2 under low-temperature environment is unclear. In this research, the sterilization effect of high-intensity ultraviolet-C (HIUVC) irradiation against SARS-CoV-2 and Staphylococcus aureus on different carriers at 4 °C and - 20 °C was investigated. The results showed that dose of 15.3 mJ/cm2 achieved more than 3 log reduction of SARS-CoV-2 on gauze at 4 °C and - 20 °C. The vulnerability of coronavirus to HIUVC under - 20 °C was not significantly different than those under 4 °C. Four models including Weibull, biphasic, log-linear tail and log linear were used to fit the survival curves of SARS-CoV-2 and Staphylococcus aureus. The biphasic model fitted best with R2 ranging from 0.9325 to 0.9878. Moreover, the HIUVC sterilization correlation between SARS-CoV-2 and Staphylococcus aureus was established. This paper provides data support for the employment of HIUVC under low-temperature environment. Also, it provides a method of using Staphylococcus aureus as a marker to evaluate the sterilization effect of cold chain sterilization equipment.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Temperatura , Refrigeração , Raios Ultravioleta
6.
J Med Virol ; 95(3): e28657, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36912367

RESUMO

Novel immune escape variants have emerged as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread worldwide. Many of the variants cause breakthrough infections in vaccinated populations, posing great challenges to current antiviral strategies targeting the immunodominance of the receptor-binding domain within the spike protein. Here, we found that a novel broadly neutralizing monoclonal antibody (mAb), G5, provided efficient protection against SARS-CoV-2 variants of concern (VOCs) in vitro and in vivo. A single dose of mAb G5 could significantly inhibit the viral burden in mice challenged with the mouse-adapted SARS-CoV-2 or SARS-CoV-2 Omicron BA.1 variant, as well as the body weight loss and cytokine release induced by mouse-adapted SARS-CoV-2. The refined epitope recognized by mAb G5 was identified as 1148 FKEELDKYF1156 in the stem helix of subunit S2. In addition, a human-mouse chimeric mAb was generated based on the variable region of heavy chain and VL genes of mAb G5. Our study provides a broad antibody drug candidate against SARS-CoV-2 VOCs and reveals a novel target for developing pan-SARS-CoV-2 vaccines.


Assuntos
Anticorpos Monoclonais , COVID-19 , Humanos , Animais , Camundongos , Anticorpos Monoclonais/uso terapêutico , Vacinas contra COVID-19 , SARS-CoV-2/genética , Imunossupressores , Glicoproteína da Espícula de Coronavírus/genética , Anticorpos Neutralizantes , Anticorpos Antivirais/uso terapêutico
7.
Microbiol Spectr ; 10(5): e0231522, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36102524

RESUMO

Six highly pathogenic avian influenza (HPAI) H5N1 viruses (clade 2.3.4.4b) were detected in migratory birds in Hubei Province in November 2021. Phylogenetic analysis indicated that the viruses in the study included two different reassortants between H5N1 viruses that were circulating in Eurasia and low-pathogenic avian influenza viruses (LPAIVs). Several amino acid substitutions that contributed to the enhanced replication or virulence in mammals were observed in these viruses, suggesting a potential threat of the H5N1 viruses to human health. IMPORTANCE Here, we obtained the whole-genomes of six H5N1 viruses from dead or rescued wild birds in Hubei Province. These viruses were divided into two genotypes and had different evolutionary trajectories from previously reported H5N1 viruses in China. Extensive reassortment events between high-pathogenic (HP) and low-pathogenic (LP) avian influenza viruses (AIVs) were observed in these viruses. Moreover, a key amino acid analysis also suggests a potential threat of H5N1 viruses to public health. Our work explored the prevalent patterns of H5N1 viruses in wild birds and replenished the viral population data of H5N1 viruses in central China.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Influenza Aviária , Animais , Humanos , Virus da Influenza A Subtipo H5N1/genética , Influenza Aviária/epidemiologia , Filogenia , Vírus da Influenza A/genética , Aves , Animais Selvagens , Mamíferos , China/epidemiologia , Aminoácidos
8.
J Virol ; 96(12): e0049422, 2022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35604143

RESUMO

G protein subunit ß1 (GNB1), the beta subunit of the G protein family, plays an important role in regulating transmembrane signal transduction. Although a recent study has demonstrated that GNB1 can bind the matrix protein 1 (M1) to facilitate M1 transport to budding sites and promote the release of progeny influenza A virus (IAV), whether the GNB1 protein has other functions in IAV replication requires further study. Here, we found that GNB1 promoted IAV replication, as virus yield decreased in GNB1 knockdown or knockout cells. GNB1 interacted with polymerase subunits PB2, PB1, and PA. Overexpressed GNB1 facilitated PB2 binding to importin α3, α5, and α7 promoting the nuclear import of PB2, enhancing viral RNA synthesis and polymerase activity. Altogether, our results demonstrated that GNB1 positively regulates virus replication by interacting with polymerase subunits and facilitating the nuclear import of PB2, which provide novel insights into the molecular mechanism of IAV. IMPORTANCE Until now, there has been only one article on the role of GNB1 in IAV budding. No study has investigated the role of GNB1 in IAV replication. In this study, our research demonstrated that GNB1 could increase the interaction between PB2 and the importin α isoform and mediate the nuclear import of PB2. Therefore, GNB1 could promote viral replication and transcription. Our results provide a better understanding of the molecular mechanisms of viral replication and provide potential antiviral drug targets.


Assuntos
Transporte Ativo do Núcleo Celular , Subunidades beta da Proteína de Ligação ao GTP , Vírus da Influenza A , Influenza Humana , Proteínas Virais , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Influenza Humana/genética , Carioferinas/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
10.
mBio ; 13(3): e0061522, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35638835

RESUMO

Upon influenza A virus (IAV) infection, the IAV progeny ribonucleoprotein complex, with a defective viral genome, is sensed by DNA-dependent activator of interferon-regulatory factor (DAI). DAI initiates the recruitment of an array of proteins to form a multiprotein platform (PANoptosome), which triggers apoptosis, necroptosis, and pyroptosis during IAV infection. However, the mechanisms mediating the assembly of the PANoptosome are unclear. Here, we identified a scaffold protein, sperm-associated antigen 9 (SPAG9), which could interact with DAI to promote cell death during IAV infection. We further demonstrated that the cell death enhanced by SPAG9 was achieved through the DAI/SPAG9/c-Jun N-terminal kinase (JNK) axis, which could promote IAV-induced DAI-mediated cell death, including apoptosis, necroptosis, and pyroptosis. Our data further showed that the DAI/SPAG9/JNK signaling pathway enhanced the interactions among receptor-interacting serine/threonine kinase 1 (RIPK1), RIPK3, and DAI, thereby promoting IAV-induced PANoptosome formation. Overall, our study for the first time revealed a feed-forward circuit signaling pathway that enhanced IAV-induced DAI-mediated cell death, provided insights into the molecular mechanisms of cell death, and established therapeutic targets to address infectious and inflammatory diseases. IMPORTANCE Upon influenza A virus (IAV) infection, DAI is activated, recruits downstream proteins to assemble a multiprotein platform (PANoptosome), and then triggers cell death. Until now, the protein composition and assembly mechanism of the PANoptosome during IAV infection had not been elucidated. Using proximity labeling and mass spectrometry technology, we identified SPAG9 as a novel component of the PANoptosome and confirmed that SPAG9 promotes IAV-induced cell death by enhancing the interaction among RIPK1, RIPK3, and DAI. Our study will broaden the knowledge of the molecular mechanisms of cell death.


Assuntos
Vírus da Influenza A , Influenza Humana , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Morte Celular/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sêmen/metabolismo , Transdução de Sinais/genética
12.
J Nanobiotechnology ; 19(1): 295, 2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34583708

RESUMO

Fluorescent labeling and dynamic tracking is a powerful tool for exploring virus infection mechanisms. However, for small-sized viruses, virus tracking studies are usually hindered by a lack of appropriate labeling methods that do not dampen virus yield or infectivity. Here, we report a universal strategy for labeling viruses with chemical dyes and Quantum dots (QDs). Enterovirus 71 (EV71) was produced in a cell line that stably expresses a mutant methionyl-tRNA synthetase (MetRS), which can charge azidonorleucine (ANL) to the methionine sites of viral proteins during translation. Then, the ANL-containing virus was easily labeled with DBCO-AF647 and DBCO-QDs. The labeled virus shows sufficient yield and no obvious decrease in infectivity and can be used for imaging the virus entry process. Using the labeled EV71, different functions of scavenger receptor class B, member 2 (SCARB2), and heparan sulfate (HS) in EV71 infection were comparatively studied. The cell entry process of a strong HS-binding EV71 strain was investigated by real-time dynamic visualization of EV71-QDs in living cells. Taken together, our study described a universal biocompatible virus labeling method, visualized the dynamic viral entry process, and reported details of the receptor usage of EV71.


Assuntos
Enterovirus/metabolismo , Pontos Quânticos/química , Receptores Virais/metabolismo , Animais , Azidas , Linhagem Celular , Chlorocebus aethiops , Enterovirus/genética , Enterovirus Humano A/genética , Enterovirus Humano A/metabolismo , Células HeLa , Humanos , Norleucina/análogos & derivados , Receptores Depuradores/metabolismo , Células Vero , Proteínas Virais , Internalização do Vírus
13.
Clin Infect Dis ; 73(3): 361-366, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-32638022

RESUMO

BACKGROUND: Currently, there is no direct evidence to prove the active replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the intestinal tract and relevant pathological changes in the colon and rectum. We investigated the presence of virions and pathological changes in surgical rectal tissues of a patient with clinically confirmed coronavirus disease 2019 (COVID-19) with rectal adenocarcinoma. METHODS: The clinical data were collected during hospitalization and follow-up of this patient. Quantitative reverse transcriptase-polymerasechain reaction (RT-PCR) was performed on the rectal tissue specimens obtained from surgical resection, succus entericus and intestinal mucosa of ileostomy, and rectal mucosa during follow-up after recovery. Ultrathin sections of surgical samples were observed for SARS-CoV-2 virions using electron microscopy. Histopathological examination was performed using hematoxylin-eosin stain. Immunohistochemical analysis and immunofluorescence were carried out on rectal tissues to evaluate the distribution of SARS-CoV-2 antigen and immune cell infiltrations. RESULTS: The patient had fever and cough on day 3 postoperatively, was diagnosed with COVID-19 on day 7, and was discharged from the hospital on day 41. RNA of SARS-CoV-2 was detected in surgically resected rectal specimens but not in samples collected 37 days after discharge. Notably, coincident with rectal tissues of surgical specimens testing nucleic acid positive for SARS-CoV-2, typical coronavirus virions in rectal tissue were observed under electron microscopy. Moreover, abundant lymphocytes and macrophages (some were SARS-CoV-2 positive) infiltrating the lamina propria were found with no significant mucosal damage. CONCLUSIONS: We first report the direct evidence of active SARS-CoV-2 replication in a patient's rectum during the incubation period, which might explain SARS-CoV-2 fecal-oral transmission.


Assuntos
COVID-19 , SARS-CoV-2 , Hospitalização , Humanos , Intestinos , Alta do Paciente , RNA Viral
14.
Viruses ; 12(8)2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32823486

RESUMO

Enterovirus 71 (EV71) is the major causative pathogen of human hand, foot, and mouth disease (hHFMD) and has evolved to use various cellular receptors for infection. However, the relationship between receptor preference and EV71 virulence has not been fully revealed. By using reverse genetics, we identified that a single E98K mutation in VP1 is responsible for rapid viral replication in vitro. The E98K mutation enhanced binding of EV71-GZCII to cells in a heparan sulfate (HS)-dependent manner, and it attenuated the virulence of EV71-GZCII in BALB/c mice, indicating that the HS-binding property is negatively associated with viral virulence. HS is widely expressed in vascular endothelial cells in different mouse tissues, and weak colocalization of HS with scavenger receptor B2 (SCARB2) was detected. The cGZCII-98K virus bound more efficiently to mouse tissue homogenates, and the cGZCII-98K virus titers in mouse tissues and blood were much lower than the cGZCII virus titers. Together, these findings suggest that the enhanced adsorption of the cGZCII-98K virus, which likely occurs through HS, is unable to support the efficient replication of EV71 in vivo. Our study confirmed the role of HS-binding sites in EV71 infection and highlighted the importance of the HS receptor in EV71 pathogenesis.


Assuntos
Proteínas do Capsídeo/genética , Enterovirus Humano A/genética , Enterovirus Humano A/patogenicidade , Heparitina Sulfato/metabolismo , Mutação , Ligação Viral , Substituição de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Infecções por Enterovirus/virologia , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Células Vero , Carga Viral , Virulência , Replicação Viral
15.
Theranostics ; 10(14): 6430-6447, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32483462

RESUMO

Rationale: Zika virus (ZIKV) is a pathogenic virus known to cause a wide range of congenital abnormalities, including microcephaly, Guillain-Barre syndrome, meningoencephalitis, and other neurological complications, in humans. This study investigated the noninvasive detection of ZIKV infection in vivo, which is necessary for elucidating the virus's mechanisms of viral replication and pathogenesis, as well as to accelerate the development of anti-ZIKV therapeutic strategies. Methods: In this study, a recombinant ZIKV harbouring Nluc gene (ZIKV-Nluc) was designed, recovered, and purified. The levels of bioluminescence were directly correlated with viral loads in vitro and in vivo. The dynamics of ZIKV infection in A129 (interferon (IFN)-α/ß receptor deficient), AG6 (IFN-α/ß and IFN-γ receptor deficient), and C57BL/6 mice were characterized. Pregnant dams were infected with ZIKV-Nluc at E10 via intra footpad injection. Then, the pooled immune sera (anti-ZIKV neutralizing antibodies) #22-1 in ZIKV-Nluc virus-infected mice were visualized. Results: ZIKV-Nluc showed a high genetic stability and replicated well in cells with similar properties to the wild-type ZIKV (ZIKVwt). Striking bioluminescence signals were consistently observed in animal organs, including spleen, intestine, testis, uterus/ovary, and kidney. The ileocecal junction was found to be the crucial visceral target. Infection of pregnant dams with ZIKV-Nluc showed that ZIKV was capable of crossing the maternal-fetal barrier to infect the fetuses via vertical transmission. Furthermore, it was visualized that treatment with the pooled immune sera was found to greatly restrict the spread of the ZIKV-Nluc virus in mice. Conclusions: This study is the first to report the real-time noninvasive tracking of the progression of ZIKV invading immune-sheltered tissues and propagating vertically during pregnancy. The results demonstrate that ZIKV-Nluc represents a powerful tool for the study of the replication, dissemination, pathogenesis, and treatment of ZIKV in vitro and in vivo.


Assuntos
Medições Luminescentes/métodos , Infecção por Zika virus/diagnóstico por imagem , Estruturas Animais/diagnóstico por imagem , Estruturas Animais/imunologia , Estruturas Animais/virologia , Animais , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/farmacologia , Modelos Animais de Doenças , Feminino , Feto/diagnóstico por imagem , Feto/virologia , Transmissão Vertical de Doenças Infecciosas , Camundongos , Camundongos Endogâmicos C57BL/virologia , Gravidez , Carga Viral/efeitos dos fármacos , Carga Viral/imunologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia , Zika virus/efeitos dos fármacos , Infecção por Zika virus/imunologia
16.
Virol Sin ; 35(5): 637-650, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32472451

RESUMO

Zika virus (ZIKV) is emerging as a significant pathogen worldwide and may cause severe neurological disorders such as fetal microcephaly and Guillain-Barre syndrome. No drug or listed vaccines are currently available for preventing ZIKV infection. As a major target of neutralizing, ZIKV envelop (E) protein usually used for vaccine development. Nevertheless, the immunogenicity of ZIKV envelop (E) protein expressed by baculovirus display system has never been assessed. In this study, we reported a new strategy for surface display of ZIKV E protein by a recombinant baculovirus vector derived from Autographa californica multiple nuclear polyhedrosis virus (AcMNPV) and assessed its immunogenicity in mice. We produced recombinant fusion ZIKV E protein linked with signal peptide (SP) and transmembrane domain (TM) of AcMNPV GP64. The results showed that the recombinant protein was easy to produce by baculovirus display system. BALB/c mice immunized with this recombinant E protein developed ZIKV specific serum antibodies. The anti-E protein sera from the mice were able to effectively neutralize ZIKV in vitro. More importantly, AG6 (IFN-α/ß and IFN-γ receptor deficient) mice immunized with recombinant E protein were protected against lethal ZIKV challenge. Together, these findings demonstrated that the recombinant E protein displayed by baculovirus can be conveniently prepared and displayed good immunogenicity in immunized mice. It is a promising practical approach for prompting the development of vaccine and related immunology research.


Assuntos
Vacinas Virais , Infecção por Zika virus , Zika virus , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Baculoviridae , Chlorocebus aethiops , Camundongos , Camundongos Endogâmicos BALB C , Envelope Viral , Proteínas do Envelope Viral , Zika virus/imunologia
17.
Virulence ; 11(1): 113-131, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31957543

RESUMO

Zika virus (ZIKV) infection in the human central nervous system (CNS) causes Guillain-Barre syndrome, cerebellum deformity, and other diseases. Astrocytes are immune response cells in the CNS and an important component of the blood-brain barrier. Consequently, any damage to astrocytes facilitates the spread of ZIKV in the CNS. Connective tissue growth factor/Nephroblastoma overexpressed gene family 1 (CCN1), an important inflammatory factor secreted by astrocytes, is reported to regulate innate immunity and viral infection. However, the mechanism by which astrocyte viral infection affects CCN1 expression remains undefined. In this study, we demonstrate that ZIKV infection up-regulates CCN1 expression in astrocytes, thus promoting intracellular viral replication. Other studies revealed that the cAMP response element (CRE) in the CCN1 promoter is activated by the ZIKV NS3 protein. The cAMP-responsive element-binding protein (CREB), a transacting factor of the CRE, is also activated by NS3 or ZIKV. Furthermore,a specific inhibitor of CREB, i.e. SGC-CBP30, reduced ZIKV-induced CCN1 up-regulation and ZIKV replication. Moreover, co-immunoprecipitation, overexpression, and knockdown studies confirmed that the interaction between NS3 and the regulatory domain of CaMKIIα could activate the CREB pathway, thus resulting in the up-regulation of CCN1 expression and enhancement of virus replication. In conclusion, the findings of our investigations on the NS3-CaMKIIα-CREB-CCN1 pathway provide a foundation for understanding the infection mechanism of ZIKV in the CNS.


Assuntos
Proteína de Ligação a CREB/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteína Rica em Cisteína 61/metabolismo , Infecção por Zika virus/metabolismo , Zika virus/metabolismo , Animais , Astrócitos/virologia , Chlorocebus aethiops , Células HEK293 , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Serina Endopeptidases/metabolismo , Células Vero , Proteínas não Estruturais Virais , Proteínas Virais/metabolismo , Replicação Viral
18.
Anal Chem ; 91(23): 15099-15106, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31698906

RESUMO

Detection of viruses with high sensitivity is critical for the prevention and treatment of the related disease. Two homogeneous target-induced cascade amplification methods were proposed for the detection of enterovirus 71 and coxsackievirus B3. These methods both employ DNAzyme but differ in the way in which the DNAzyme is amplified. In the hybridization chain reaction (HCR)-based strategy, the DNAzyme is assembled by hairpin DNA strands, while in the rolling circle amplification (RCA)-based strategy, the DNAzyme is synthesized by the polymerase. On the basis of the virion structure, we investigated the effects of using only VP1-antibody or VP1-antibody and VP2-antibody on the detection. And the combination of two kinds of antibodies was found to further improve the performance of the detection. Subsequently, the simultaneous detection of EV71 and CVB3 was achieved by the RCA-based strategy. And the proposed methods were also applied in clinical samples analysis with a satisfactory result, showing great potential for applications in virus detection.


Assuntos
DNA Catalítico/biossíntese , Enterovirus Humano A/isolamento & purificação , Enterovirus Humano B/isolamento & purificação , Técnicas de Amplificação de Ácido Nucleico/métodos , Anticorpos Antivirais , DNA Catalítico/metabolismo , Humanos
19.
Viruses ; 11(1)2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30587778

RESUMO

Enterovirus A71 (EVA71) is a human enterovirus belonging to the Picornaviridae family and mostly causes hand-foot-and-mouth disease in infants. Viperin is an important interferon-stimulated gene with a broad antiviral activity against various viruses. However, the effect of viperin on human enteroviruses and the interaction mechanism between EVA71 and viperin remains elusive. Here, we confirmed the EVA71-induced expression of viperin in a mouse model and cell lines and showed that viperin upregulation by EVA71 infection occurred on both the mRNA and protein level. Viperin knockdown and overexpression in EVA71-infected cells indicated that this protein can markedly inhibit EVA71 infection. Interestingly, immunofluorescent confocal microscopy and co-immunoprecipitation assays indicated that viperin interacts and colocalizes with the EVA71 protein 2C in the endoplasmic reticulum. Furthermore, amino acids 50⁻60 in the N-terminal domain of viperin were the key residues responsible for viperin interaction with 2C. More importantly, the N-terminal domain of viperin was found responsible for inhibiting EVA71 replication. Our findings can potentially aid future research on the prevention and treatment of nervous system damage caused by EVA71 and may provide a potential target for antiviral therapy.


Assuntos
Proteínas de Transporte/metabolismo , Enterovirus/fisiologia , Interações entre Hospedeiro e Microrganismos , Proteínas/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Linhagem Celular Tumoral , Retículo Endoplasmático/química , Retículo Endoplasmático/virologia , Infecções por Enterovirus/virologia , Imunofluorescência , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Proteínas/metabolismo , RNA Mensageiro , Regulação para Cima
20.
J Neuroinflammation ; 15(1): 275, 2018 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-30241539

RESUMO

BACKGROUND: Many viruses depend on the extensive membranous network of the endoplasmic reticulum (ER) for their translation, replication, and packaging. Certain membrane modifications of the ER can be a trigger for ER stress, as well as the accumulation of viral protein in the ER by viral infection. Then, unfolded protein response (UPR) is activated to alleviate the stress. Zika virus (ZIKV) is a mosquito-borne flavivirus and its infection causes microcephaly in newborns and serious neurological complications in adults. Here, we investigated ER stress and the regulating model of UPR in ZIKV-infected neural cells in vitro and in vivo. METHODS: Mice deficient in type I and II IFN receptors were infected with ZIKV via intraperitoneal injection and the nervous tissues of the mice were assayed at 5 days post-infection. The expression of phospho-IRE1, XBP1, and ATF6 which were the key markers of ER stress were analyzed by immunohistochemistry assay in vivo. Additionally, the nuclear localization of XBP1s and ATF6n were analyzed by immunohistofluorescence. Furthermore, two representative neural cells, neuroblastoma cell line (SK-N-SH) and astrocytoma cell line (CCF-STTG1), were selected to verify the ER stress in vitro. The expression of BIP, phospho-elF2α, phospho-IRE1, and ATF6 were analyzed through western blot and the nuclear localization of XBP1s was performed by confocal immunofluorescence microscopy. RT-qPCR was also used to quantify the mRNA level of the UPR downstream genes in vitro and in vivo. RESULTS: ZIKV infection significantly upregulated the expression of ER stress markers in vitro and in vivo. Phospho-IRE1 and XBP1 expression significantly increased in the cerebellum and mesocephalon, while ATF6 expression significantly increased in the mesocephalon. ATF6n and XBP1s were translocated into the cell nucleus. The levels of BIP, ATF6, phospho-elf2α, and spliced xbp1 also significantly increased in vitro. Furthermore, the downstream genes of UPR were detected to investigate the regulating model of the UPR during ZIKV infection in vitro and in vivo. The transcriptional levels of atf4, gadd34, chop, and edem-1 in vivo and that of gadd34 and chop in vitro significantly increased. CONCLUSION: Findings in this study demonstrated that ZIKV infection activates ER stress in neural cells. The results offer clues to further study the mechanism of neuropathogenesis caused by ZIKV infection.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Infecção por Zika virus/metabolismo , Infecção por Zika virus/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular Transformada , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Regulação Viral da Expressão Gênica/genética , Regulação Viral da Expressão Gênica/fisiologia , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/virologia , Proteínas Serina-Treonina Quinases/genética , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Proteína 1 de Ligação a X-Box/genética , Zika virus/genética , Zika virus/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA