Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PLoS Pathog ; 19(3): e1011241, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36930690

RESUMEN

Dengue virus (DENV) infection can induce life-threatening dengue hemorrhagic fever/dengue shock syndrome in infected patients. DENV is a threat to global health due to its growing numbers and incidence of infection in the last 50 years. During infection, DENV expresses ten structural and nonstructural proteins modulating cell responses to benefit viral replication. However, the lack of knowledge regarding the cellular proteins and their functions in enhancing DENV pathogenesis impedes the development of antiviral drugs and therapies against fatal DENV infection. Here, we identified that integrin-linked kinase (ILK) is a novel enhancing factor for DENV infection by suppressing type I interferon (IFN) responses. Mechanistically, ILK binds DENV NS1 and NS3, activates Akt and Erk, and induces NF-κB-driven suppressor of cytokine signaling 3 (SOCS3) expression. Elevated SOCS3 in DENV-infected cells inhibits phosphorylation of STAT1/2 and expression of interferon-stimulated genes (ISGs). Inhibiting ILK, Akt, or Erk activation abrogates SOCS3 expression. In DENV-infected mice, the treatment of an ILK inhibitor significantly reduces viral loads in the brains, disease severity, and mortality rate. Collectively, our results show that ILK is a potential therapeutic target against DENV infection.


Asunto(s)
Virus del Dengue , Dengue , Interferón Tipo I , Animales , Ratones , Virus del Dengue/fisiología , Proteínas Proto-Oncogénicas c-akt , Replicación Viral , Interferón Tipo I/uso terapéutico
2.
Proc Natl Acad Sci U S A ; 117(27): 15947-15954, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32576686

RESUMEN

The cytosolic DNA sensor cGMP-AMP synthase (cGAS) synthesizes the noncanonical cyclic dinucleotide 2'3'-cGAMP to activate the adaptor protein stimulator of IFN genes (STING), thus awakening host immunity in response to DNA pathogen infection. However, dengue virus (DENV), an RNA virus without a DNA stage in its life cycle, also manipulates cGAS-STING-mediated innate immunity by proteolytic degradation of STING. Here, we found that the sensitivity of STING to DENV protease varied with different human STING haplotypes. Exogenous DNA further enhanced DENV protease's ability to interact and cleave protease-sensitive STING. DNA-enhanced STING cleavage was reduced in cGAS-knockdown cells and triggered by the cGAS product 2'3'-cGAMP. The source of DNA may not be endogenous mitochondrial DNA but rather exogenous reactivated viral DNA. Cells producing 2'3'-cGAMP by overexpressing cGAS or with DNA virus reactivation enhanced STING cleavage in neighboring cells harboring DENV protease. DENV infection reduced host innate immunity in cells with the protease-sensitive STING haplotype, whose homozygote genotype frequency was found significantly reduced in Taiwanese people with dengue fever. Therefore, the human STING genetic background and DNA pathogen coinfection may be the missing links contributing to DENV pathogenesis.


Asunto(s)
Dengue/enzimología , Endopeptidasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Nucleótidos Cíclicos/metabolismo , Células A549 , ADN Viral/genética , Dengue/inmunología , Endopeptidasas/genética , Haplotipos , Humanos , Evasión Inmune , Inmunidad Innata , Nucleótidos Cíclicos/genética
3.
J Biomed Sci ; 29(1): 37, 2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35681239

RESUMEN

BACKGROUND: Calls for the coronavirus to be treated as an endemic illness, such as the flu, are increasing. After achieving high coverage of COVID-19 vaccination, therapeutic drugs have become important for future SARS-CoV-2 variant outbreaks. Although many monoclonal antibodies have been approved for emergency use as treatments for SARS-CoV-2 infection, some monoclonal antibodies are not authorized for variant treatment. Broad-spectrum monoclonal antibodies are unmet medical needs. METHODS: We used a DNA prime-protein boost approach to generate high-quality monoclonal antibodies. A standard ELISA was employed for the primary screen, and spike protein-human angiotensin-converting enzyme 2 blocking assays were used for the secondary screen. The top 5 blocking clones were selected for further characterization, including binding ability, neutralization potency, and epitope mapping. The therapeutic effects of the best monoclonal antibody against SARS-CoV-2 infection were evaluated in a hamster infection model. RESULTS: Several monoclonal antibodies were selected that neutralize different SARS-CoV-2 variants of concern (VOCs). These VOCs include Alpha, Beta, Gamma, Delta, Kappa and Lambda variants. The high neutralizing antibody titers against the Beta variant would be important to treat Beta-like variants. Among these monoclonal antibodies, mAb-S5 displays the best potency in terms of binding affinity and neutralizing capacity. Importantly, mAb-S5 protects animals from SARS-CoV-2 challenge, including the Wuhan strain, D614G, Alpha and Delta variants, although mAb-S5 exhibits decreased neutralization potency against the Delta variant. Furthermore, the identified neutralizing epitopes of monoclonal antibodies are all located in the receptor-binding domain (RBD) of the spike protein but in different regions. CONCLUSIONS: Our approach generates high-potency monoclonal antibodies against a broad spectrum of VOCs. Multiple monoclonal antibody combinations may be the best strategy to treat future SARS-CoV-2 variant outbreaks.


Asunto(s)
Anticuerpos Monoclonales , Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Vacunas contra la COVID-19 , Cricetinae , Humanos , Glicoproteína de la Espiga del Coronavirus/genética
4.
J Immunol ; 203(7): 1909-1917, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31451673

RESUMEN

Dengue virus (DENV) causes a range of illness, including dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. DENV nonstructural protein (NS) 1 has been considered to be a desirable vaccine candidate for its ability to induce Ab and complement-dependent cytolysis of DENV-infected cells as well as to block the pathogenic effects of NS1. However a potential drawback of NS1 as a vaccine is that anti-DENV NS1 Abs can lead to endothelial cell damage and platelet dysfunction by antigenic cross-reactivity. Therefore, we modified the DENV NS1 by replacing the C-terminal cross-reactive epitopes with the corresponding region of Japanese encephalitis virus NS1 to generate a chimeric DJ NS1 protein. Active immunization with DJ NS1 induced a strong Ab response. To enhance cellular immunity, we further combined DJ NS1 with DENV NS3 to immunize mice and showed activation of Ag-specific CD4+ and CD8+ T cells in addition to Ab responses. We further detected NS3-specific CTL activities as well as CD107a expression of effector cells. Importantly, the protective effects attributed by DJ NS1 and NS3 immunization were demonstrated in a DENV-infected mouse model by reduced viral titers, soluble NS1 levels, mouse tail bleeding time, and vascular leakage at skin injection sites. Collectively, the results from this study reveal the humoral and cellular immune responses and the protective effects conferred by DJ NS1 and NS3 immunization in the mouse model of DENV infection and provide a potential strategy for dengue vaccine design.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Dengue/inmunología , Virus del Dengue/inmunología , Dengue/prevención & control , Inmunidad Celular , Inmunización , Proteínas no Estructurales Virales/inmunología , Animales , Reacciones Cruzadas , Dengue/inmunología , Dengue/patología , Epítopos/inmunología , Masculino , Ratones
5.
Int J Mol Sci ; 22(16)2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34445789

RESUMEN

The SARS-CoV-2 pseudovirus is a commonly used strategy that mimics certain biological functions of the authentic virus by relying on biological legitimacy at the molecular level. Despite the fact that spike (S), envelope (E), and membrane (M) proteins together wrap up the SARS-CoV-2 virion, most of the reported pseudotype viruses consist of only the S protein. Here, we report that the presence of E and M increased the virion infectivity by promoting the S protein priming. The S, E, and M (SEM)-coated pseudovirion is spherical, containing crown-like spikes on the surface. Both S and SEM pseudoviruses packaged the same amounts of viral RNA, but the SEM virus bound more efficiently to cells stably expressing the viral receptor human angiotensin-converting enzyme II (hACE2) and became more infectious. Using this SEM pseudovirus, we examined the infectivity and antigenic properties of the natural SARS-CoV-2 variants. We showed that some variants have higher infectivity than the original virus and that some render the neutralizing plasma with lower potency. These studies thus revealed possible mechanisms of the dissemination advantage of these variants. Hence, the SEM pseudovirion provides a useful tool to evaluate the viral infectivity and capability of convalescent sera in neutralizing specific SARS-CoV-2 S dominant variants.


Asunto(s)
Anticuerpos Antivirales/metabolismo , COVID-19/inmunología , Proteínas de la Envoltura de Coronavirus/metabolismo , SARS-CoV-2/patogenicidad , Proteínas de la Matriz Viral/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Antivirales/inmunología , COVID-19/sangre , COVID-19/virología , Línea Celular , Proteínas de la Envoltura de Coronavirus/genética , Proteínas de la Envoltura de Coronavirus/inmunología , Proteínas de la Envoltura de Coronavirus/ultraestructura , Cricetinae , Humanos , Microscopía Electrónica de Transmisión , Mutación , Pruebas de Neutralización , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Proteínas de la Matriz Viral/ultraestructura , Virión/genética , Virión/inmunología , Virión/metabolismo , Virión/ultraestructura
6.
Int J Mol Sci ; 21(24)2020 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-33352639

RESUMEN

Dengue virus (DENV) infection is a significant public health threat in tropical and subtropical regions; however, there is no specific antiviral drug. Accumulated studies have revealed that DENV infection induces several cellular responses, including autophagy and apoptosis. The crosstalk between autophagy and apoptosis is associated with the interactions among components of these two pathways, such as apoptotic caspase-mediated cleavage of autophagy-related proteins. Here, we show that DENV-induced autophagy inhibits early cell apoptosis and hence enhances DENV replication. Later, the apoptotic activities are elevated to suppress autophagy through cleavage of Beclin-1, an essential autophagy-related protein. Inhibition of cleavage of Beclin-1 by a pan-caspase inhibitor, Z-VAD, increases both autophagy and viral replication. Regarding the mechanism, we further found that DENV nonstructural protein 1 (NS1) is able to interact with Beclin-1 during DENV infection. The interaction between Beclin-1 and NS1 attenuates Beclin-1 cleavage and facilitates autophagy to prevent cell apoptosis. Our study suggests a novel mechanism whereby NS1 preserves Beclin-1 for maintaining autophagy to antagonize early cell apoptosis; however, elevated caspases trigger apoptosis by degrading Beclin-1 in the late stage of infection. These findings suggest implications for anti-DENV drug design.


Asunto(s)
Beclina-1/metabolismo , Caspasas/metabolismo , Virus del Dengue/aislamiento & purificación , Dengue/patología , Dengue/virología , Proteínas no Estructurales Virales/metabolismo , Células A549 , Aedes , Animales , Autofagia , Dengue/metabolismo , Humanos
7.
J Virol ; 90(9): 4308-4319, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26889037

RESUMEN

UNLABELLED: Small ubiquitin-like modifier (SUMO) participates in a reversible posttranslational modification process (SUMOylation) that regulates a wide variety of cellular processes and plays important roles for numerous viruses during infection. However, the roles of viral protein SUMOylation in dengue virus (DENV) infection have not been elucidated. In this study, we found that the SUMOylation pathway was involved in the DENV life cycle, since DENV replication was reduced by silencing the cellular gene Ubc9, which encodes the sole E2-conjugating enzyme required for SUMOylation. By in vivo and in vitro SUMOylation assays, the DENV NS5 protein was identified as an authentic SUMO-targeted protein. By expressing various NS5 mutants, we found that the SUMO acceptor sites are located in the N-terminal domain of NS5 and that a putative SUMO-interacting motif (SIM) of this domain is crucial for its SUMOylation. A DENV replicon harboring the SUMOylation-defective SIM mutant showed a severe defect in viral RNA replication, supporting the notion that NS5 SUMOylation is required for DENV replication. SUMOylation-defective mutants also failed to suppress the induction of STAT2-mediated host antiviral interferon signaling. Furthermore, the SUMOylation of NS5 significantly increased the stability of NS5 protein, which could account for most of the biological functions of SUMOylated NS5. Collectively, these findings suggest that the SUMOylation of DENV NS5 is one of the mechanisms regulating DENV replication. IMPORTANCE: SUMOylation is a common posttranslational modification that regulates cellular protein functions but has not been reported in the proteins of dengue virus. Here, we found that the replicase of DENV, nonstructural protein 5 (NS5), can be SUMOylated. It is well known that providing RNA-dependent RNA polymerase activity and antagonizing host antiviral IFN signaling are a "double indemnity" of NS5 to support DENV replication. Without SUMOylation, NS5 fails to maintain its protein stability, which consequently disrupts its function in viral RNA replication and innate immunity antagonism. DENV threatens billions of people worldwide, but no licensed vaccine or specific therapeutics are currently available. Thus, our findings suggest that rather than specifically targeting NS5 enzyme activity, NS5 protein stability is a novel drug target on the growing list of anti-DENV strategies.


Asunto(s)
Virus del Dengue/fisiología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Dengue/genética , Dengue/metabolismo , Dengue/virología , Silenciador del Gen , Humanos , Datos de Secuencia Molecular , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Interferencia de ARN , Alineación de Secuencia , Sumoilación , Enzimas Ubiquitina-Conjugadoras/genética , Proteínas no Estructurales Virales/química
8.
PLoS Pathog ; 11(12): e1005350, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26717518

RESUMEN

Mitochondria are highly dynamic subcellular organelles participating in many signaling pathways such as antiviral innate immunity and cell death cascades. Here we found that mitochondrial fusion was impaired in dengue virus (DENV) infected cells. Two mitofusins (MFN1 and MFN2), which mediate mitochondrial fusion and participate in the proper function of mitochondria, were cleaved by DENV protease NS2B3. By knockdown and overexpression approaches, these two MFNs showed diverse functions in DENV infection. MFN1 was required for efficient antiviral retinoic acid-inducible gene I-like receptor signaling to suppress DENV replication, while MFN2 participated in maintaining mitochondrial membrane potential (MMP) to attenuate DENV-induced cell death. Cleaving MFN1 and MFN2 by DENV protease suppressed mitochondrial fusion and deteriorated DENV-induced cytopathic effects through subverting interferon production and facilitating MMP disruption. Thus, MFNs participate in host defense against DENV infection by promoting the antiviral response and cell survival, and DENV regulates mitochondrial morphology by cleaving MFNs to manipulate the outcome of infection.


Asunto(s)
Virus del Dengue/metabolismo , Dengue/metabolismo , GTP Fosfohidrolasas/metabolismo , Interacciones Huésped-Parásitos/inmunología , Dinámicas Mitocondriales/fisiología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Western Blotting , Citometría de Flujo , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Replicación Viral/inmunología
9.
J Immunol ; 191(6): 3328-36, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23940278

RESUMEN

The IFN immune system comprises type I, II, and III IFNs, signals through the JAK-STAT pathway, and plays central roles in host defense against viral infection. Posttranslational modifications such as ubiquitination regulate diverse molecules in the IFN pathway. To search for the deubiquitinating enzymes (DUBs) involved in the antiviral activity of IFN, we used RNA interference screening to identify a human DUB, ubiquitin-specific protease (USP) 13, whose expression modulates the antiviral activity of IFN-α against dengue virus serotype 2 (DEN-2). The signaling events and anti-DEN-2 activities of IFN-α and IFN-γ were reduced in cells with USP13 knockdown but enhanced with USP13 overexpression. USP13 may regulate STAT1 protein because the protein level and stability of STAT1 were increased with USP13 overexpression. Furthermore, STAT1 ubiquitination was reduced in cells with USP13 overexpression and increased with USP13 knockdown regardless of with or without IFN-α treatment. Thus, USP13 positively regulates type I and type II IFN signaling by deubiquitinating and stabilizing STAT1 protein. Overall, to our knowledge, USP13 is the first DUB identified to modulate STAT1 and play a role in the antiviral activity of IFN against DEN-2 replication.


Asunto(s)
Virus del Dengue/inmunología , Endopeptidasas/metabolismo , Interferones/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/inmunología , Western Blotting , Endopeptidasas/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Interferones/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/inmunología , Proteasas Ubiquitina-Específicas
10.
Mediators Inflamm ; 2015: 274025, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26199460

RESUMEN

Infection with dengue virus (DENV) causes an increase in proinflammatory responses, such as nitric oxide (NO) generation and TNF-α expression; however, the molecular mechanism underlying this inflammatory activation remains undefined, although the activation of the transcription factor NF-κB is generally involved. In addition to TNF-α production in DENV-infected murine macrophage RAW264.7 cells, inducible NO synthase was transcriptionally and posttranslationally elevated and accompanied by NO generation. NF-κB is known to be activated by DENV infection. Pharmacologically inhibiting NF-κB activation abolishes iNOS/NO biosynthesis and TNF-α production. With inhibition, the potential role of NF-κB in oxidative signaling regulation was prevented during DENV infection. Heat-inactivated DENV failed to cause the identified inflammatory responses. Pharmacological inhibition of TLR3 partly decreased NF-κB activation; however, it effectively abolished inducible iNOS/NO biosynthesis but did not inhibit TNF-α production. In contrast to TLR3, viral protein NS2B3 also independently contributed to NF-κB activation to regulate TNF-α production. These results show the distinct pathways for NF-κB activation caused by DENV infection individually for the regulation of iNOS/NO and TNF-α expression.


Asunto(s)
Virus del Dengue/patogenicidad , Macrófagos/metabolismo , Macrófagos/virología , FN-kappa B/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Ácidos Cafeicos/farmacología , Línea Celular , Macrófagos/efectos de los fármacos , Ratones , Óxido Nítrico/metabolismo , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Transducción de Señal/efectos de los fármacos
11.
PLoS Pathog ; 8(6): e1002780, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761576

RESUMEN

Dengue is one of the most important arboviral diseases caused by infection of four serotypes of dengue virus (DEN). We found that activation of interferon regulatory factor 3 (IRF3) triggered by viral infection and by foreign DNA and RNA stimulation was blocked by DEN-encoded NS2B3 through a protease-dependent mechanism. The key adaptor protein in type I interferon pathway, human mediator of IRF3 activation (MITA) but not the murine homologue MPYS, was cleaved in cells infected with DEN-1 or DEN-2 and with expression of the enzymatically active protease NS2B3. The cleavage site of MITA was mapped to LRR↓(96)G and the function of MITA was suppressed by dengue protease. DEN replication was reduced with overexpression of MPYS but not with MITA, while DEN replication was enhanced by MPYS knockdown, indicating an antiviral role of MITA/MPYS against DEN infection. The involvement of MITA in DEN-triggered innate immune response was evidenced by reduction of IRF3 activation and IFN induction in cells with MITA knockdown upon DEN-2 infection. NS2B3 physically interacted with MITA, and the interaction and cleavage of MITA could be further enhanced by poly(dA:dT) stimulation. Thus, we identified MITA as a novel host target of DEN protease and provide the molecular mechanism of how DEN subverts the host innate immunity.


Asunto(s)
Virus del Dengue/metabolismo , Dengue/metabolismo , Inmunidad Innata/inmunología , Proteínas de la Membrana/metabolismo , Animales , Dengue/genética , Dengue/inmunología , Virus del Dengue/genética , Virus del Dengue/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunidad Innata/genética , Immunoblotting , Inmunoprecipitación , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Péptido Hidrolasas/genética , Péptido Hidrolasas/inmunología , Péptido Hidrolasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
12.
Nat Commun ; 15(1): 6607, 2024 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-39098891

RESUMEN

Delivering synthetic protein-coding RNA bypassing the DNA stage for ectopic protein functioning is a novel therapeutic strategy. Joining the linear RNA head-to-tail covalently could be a state-of-the-art strategy for functioning longer. Here we enroll a cis-acting ligase ribozyme (RzL) to generate circular RNA (circRNA) in vitro for ectopic protein expression. The RNA circularization is confirmed by masking the 5' phosphate group, resisting exonuclease RNase R digestion, failing for further tailing, and sequencing the RT-PCR products of the joined region. Interestingly, one internal ribosome entry site (IRES) renders circRNA translation competent, but two IRES in cis, not trans, hamper the translation. The circRNA with highly potent in translation is conferred for antiviral functioning. Accompanying specific guided RNA, a circRNA expressing ribonuclease Cas13 shows excellent potential against the corresponding RNA virus, further extending circRNA functioning in its growing list of applications.


Asunto(s)
ARN Catalítico , ARN Circular , ARN Circular/metabolismo , ARN Circular/genética , ARN Catalítico/metabolismo , ARN Catalítico/genética , Humanos , Sitios Internos de Entrada al Ribosoma , Biosíntesis de Proteínas , ARN/metabolismo , ARN/genética , Células HEK293 , Exorribonucleasas
13.
J Virol ; 86(19): 10347-58, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22787234

RESUMEN

Japanese encephalitis virus (JEV) is an enveloped flavivirus with a single-stranded, positive-sense RNA genome encoding three structural and seven nonstructural proteins. To date, the role of JEV nonstructural protein 2A (NS2A) in the viral life cycle is largely unknown. The interferon (IFN)-induced double-stranded RNA (dsRNA)-activated protein kinase (PKR) phosphorylates the eukaryotic translation initiation factor 2α subunit (eIF2α) after sensing viral RNA and results in global translation arrest as an important host antiviral defense response. In this study, we found that JEV NS2A could antagonize PKR-mediated growth inhibition in a galactose-inducible PKR-expressing yeast system. In human cells, PKR activation, eIF2α phosphorylation, and the subsequent translational inhibition and cell death triggered by dsRNA and IFN-α were also repressed by JEV NS2A. Moreover, among the four eIF2α kinases, NS2A specifically blocked the eIF2α phosphorylation mediated by PKR and attenuated the PKR-promoted cell death induced by the chemotherapeutic drug doxorubicin. A single point mutation of NS2A residue 33 from Thr to Ile (T33I) abolished the anti-PKR potential of JEV NS2A. The recombinant JEV mutant carrying the NS2A-T33I mutation showed reduced in vitro growth and in vivo virulence phenotypes. Thus, JEV NS2A has a novel function in blocking the host antiviral response of PKR during JEV infection.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/metabolismo , Regulación Viral de la Expresión Génica , ARN Bicatenario/metabolismo , Proteínas Virales/química , eIF-2 Quinasa/antagonistas & inhibidores , Animales , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Activación Enzimática , Genes Reporteros , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación , Fenotipo , Plásmidos/metabolismo , Biosíntesis de Proteínas , Saccharomyces cerevisiae/metabolismo , Proteínas Virales/metabolismo , eIF-2 Quinasa/metabolismo
14.
Mol Ther Oncolytics ; 28: 104-117, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36699618

RESUMEN

Glioblastoma (GBM) is the most common aggressive malignant brain cancer and is chemo- and radioresistant, with poor therapeutic outcomes. The "double-edged sword" of virus-induced cell death could be a potential solution if the oncolytic virus specifically kills cancer cells but spares normal ones. Zika virus (ZIKV) has been defined as a prospective oncolytic virus by selectively targeting GBM cells, but unclear understanding of how ZIKV kills GBM and the consequences hinders its application. Here, we found that the cellular gasdermin D (GSDMD) is required for the efficient death of a human GBM cell line caused by ZIKV infection. The ZIKV protease specifically cleaves human GSDMD to activate caspase-independent pyroptosis, harming both viral protease-harboring and naive neighboring cells. Analyzing human GSDMD variants showed that most people were susceptible to ZIKV-induced cytotoxicity, except for those with variants that resisted ZIKV cleavage or were defective in oligomerizing the N terminus GSDMD cleavage product. Consistently, ZIKV-induced secretion of the pro-inflammatory cytokine interleukin-1ß and cytolytic activity were both stopped by a small-molecule inhibitor targeting GSDMD oligomerization. Thus, potential ZIKV oncolytic therapy for GBM would depend on the patient's GSDMD genetic background and could be abolished by GSDMD inhibitors if required.

15.
Cell Microbiol ; 13(9): 1358-70, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21707907

RESUMEN

Japanese encephalitis virus (JEV), a mosquito-borne flavivirus, causes acute encephalitis with high mortality in humans. We used a pair of virulent (RP-9) and attenuated (RP-2ms) variants of JEV to pull down the cell surface molecules bound with JEV particle; their identities were revealed by LC-MS/MS analysis. One major protein bound with RP-9 and weakly with RP-2ms was identified as the intermediate filament protein vimentin. Infection of RP-9 but not that of RP-2ms was blocked by anti-vimentin antibodies and by recombinant-expressed vimentin proteins. Knockdown of vimentin expression reduced the levels of viral binding and viral production of RP-9, but not that of RP-2ms. The different vimentin dependency for JEV infection could be attributed to the major structural envelope protein, as the recombinant RP-9 with an E-E138K mutation became resistant to anti-vimentin blockage. Furthermore, RP-2ms mainly depended on cell surface glycosaminoglycans for viral binding and it became vimentin-dependent only when binding to glycosaminoglycans was blocked. Thus, we suggest that vimentin contributes to virulent JEV infection and might be a new target to intervene in this deadly infection.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/metabolismo , Vimentina/metabolismo , Animales , Línea Celular Tumoral , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Glicosaminoglicanos/metabolismo , Humanos , Ratones , Unión Proteica , Vimentina/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virulencia
16.
Sci Rep ; 12(1): 3297, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35228672

RESUMEN

Preterm delivery of low-birth weight infants is considered a leading cause of morbidity and mortality among neonates. Various studies have reported a positive correlation between periodontal disease (PD) and premature birth (PB) and yet no population-based study has assessed the impact of PD severity and treatments on premature birth. This cohort study used Taiwan's national medical records (1999-2012, included 1,757,774 pregnant women) to investigate the association between PD severity and PB. Women with PD during the 2-year period prior for giving birth were more likely to have PB (11.38%) than those without PD (10.56%; p < 0.001). After variables adjustment, the advanced PD group had OR of 1.09 (95% CI 1.07-1.11) for PB, the mild PD group had OR of 1.05 (95% CI 1.04-1.06), while no-PD group had OR of 1. Increased PD severity was related to higher risk of PB. When stratified by age, the highest ORs for PB were those aged from 31 to 35 years in both mild PD group (OR = 1.09, 95% CI 1.07-1.11) and advanced PD group (OR = 1.13, 95% CI 1.09-1.17). Improving periodontal health before or during pregnancy may prevent or reduce the occurrence of adverse pregnancy outcomes and therefore maternal and perinatal morbidity and mortality.


Asunto(s)
Enfermedades Periodontales , Complicaciones del Embarazo , Nacimiento Prematuro , Adulto , Estudios de Cohortes , Femenino , Humanos , Lactante , Recién Nacido , Enfermedades Periodontales/epidemiología , Embarazo , Complicaciones del Embarazo/epidemiología , Resultado del Embarazo/epidemiología , Nacimiento Prematuro/epidemiología , Nacimiento Prematuro/prevención & control , Taiwán/epidemiología
17.
JCI Insight ; 7(21)2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36125898

RESUMEN

Diabetes mellitus (DM) is highly comorbid with severe dengue diseases; however, the underlying mechanisms are unclear. Patients with DM have a 1.61-fold increased risk of developing dengue hemorrhagic fever. In search of host factors involved in dengue virus (DENV) infection, we used high-glucose (HG) treatment and showed that HG increased viral protein expression and virion release but had no effects on the early stages of viral infection. After HG stimulation, DENV-firefly luciferase-transfected assay and cellular replicon-based assay indicated increased viral translation, whereas using the glucose uptake inhibitor phloretin blocked this effect. HG treatment increased the translational factor poly(A)-binding protein (PABP) in a glucose transporter-associated, PI3K/AKT-regulated manner. Silencing PABP significantly decreased HG-prompted virion production. HG enhanced the formation of the PABP-eukaryotic translation initiation factor 4G complex, which is regulated by protein-disulfide isomerase. Hyperglycemia increased PABP expression, mortality rate, viral protein expression, and viral loads in streptozotocin-induced DM mice. Overall, hyperglycemic stress facilitates DENV infection by strengthening PABP-mediated viral translation.


Asunto(s)
Dengue , Hiperglucemia , Animales , Ratones , Biosíntesis de Proteínas , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión a Poli(A)/genética , Proteínas de Unión a Poli(A)/metabolismo , Proteínas Virales/metabolismo , Hiperglucemia/complicaciones
18.
NPJ Vaccines ; 7(1): 60, 2022 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-35662254

RESUMEN

A major challenge in the use of DNA vaccines is efficient DNA delivery in vivo. Establishing a safe and efficient electric transfer method is the key to developing rapid DNA vaccines against emerging infectious diseases. To overcome the complexity of designing new electric transfer machines for DNA delivery, a clinically approved electric transfer machine could be considered as an alternative. Here, we report an electroacupuncture machine-based method for DNA vaccine delivery after intramuscular injection of the COVID-19 DNA vaccine. The S gene of SARS-CoV-2 in the pVAX1 plasmid (pSARS2-S) was used as an antigen in this study. We optimized the clinically used electroacupuncture machine settings for efficient induction of the neutralizing antibody titer after intramuscular injection of pSARS2-S in mice. We found that pSARS2-S immunization at 40 Vpp for 3-5 s could induce high neutralizing antibody titers and Th1-biased immune responses. IFN-γ/TNF-α-secreting CD4+ and CD8+ T cells were also observed in the DNA vaccination group but not in the recombinant protein vaccination group. T-cell epitope mapping shows that the major reactive epitopes were located in the N-terminal domain (a.a. 261-285) and receptor-binding domain (a.a. 352-363). Importantly, pSARS2-S immunization in hamsters could induce protective immunity against SARS-CoV-2 challenge in vivo. In the preclinical toxicology study, blood biochemistry, hematology, and DNA persistence analysis reveal that the DNA delivery method is safe. Furthermore, the raised antisera could also cross-neutralize different variants of concern. These findings suggest that DNA vaccination using an electroacupuncture machine is feasible for use in humans in the future.

19.
JCI Insight ; 7(8)2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35290246

RESUMEN

Most therapeutic mAbs target the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2. Unfortunately, the RBD is a hot spot for mutations in SARS-CoV-2 variants, which will lead to loss of the neutralizing function of current therapeutic mAbs. Universal mAbs for different variants are necessary. We identified mAbs that recognized the S2 region of the spike protein, which is identical in different variants. The mAbs could neutralize SARS-CoV-2 infection and protect animals from SARS-CoV-2 challenge. After cloning the variable region of the light chain and heavy chain, the variable region sequences were humanized to select a high-affinity humanized mAb, hMab5.17. hMab5.17 protected animals from SARS-CoV-2 challenge and neutralized SARS-CoV-2 variant infection. We further identified the linear epitope of the mAb, which is not mutated in any variant of concern. These data suggest that a mAb recognizing the S2 region of the spike protein will be a potential universal therapeutic mAb for COVID-19.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales , Humanos , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética
20.
iScience ; 25(8): 104709, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35813875

RESUMEN

Post-translational modifications (PTMs), such as glycosylation and palmitoylation, are critical to protein folding, stability, intracellular trafficking, and function. Understanding regulation of PTMs of SARS-CoV-2 spike (S) protein could help the therapeutic drug design. Herein, the VSV vector was used to produce SARS-CoV-2 S pseudoviruses to examine the roles of the 611LYQD614 and cysteine-rich motifs in S protein maturation and virus infectivity. Our results show that 611LY612 mutation alters S protein intracellular trafficking and reduces cell surface expression level. It also changes S protein glycosylation pattern and decreases pseudovirus infectivity. The S protein contains four cysteine-rich clusters with clusters I and II as the main palmitoylation sites. Mutations of clusters I and II disrupt S protein trafficking from ER-to-Golgi, suppress pseudovirus production, and reduce spike-mediated membrane fusion activity. Taken together, glycosylation and palmitoylation orchestrate the S protein maturation processing and are critical for S protein-mediated membrane fusion and infection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA