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












Base de datos
Intervalo de año de publicación
2.
EMBO Mol Med ; 16(6): 1310-1323, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38745062

RESUMEN

Vaccination has successfully controlled several infectious diseases although better vaccines remain desirable. Host response to vaccination studies have identified correlates of vaccine immunogenicity that could be useful to guide development and selection of future vaccines. However, it remains unclear whether these findings represent mere statistical correlations or reflect functional associations with vaccine immunogenicity. Functional associations, rather than statistical correlates, would offer mechanistic insights into vaccine-induced adaptive immunity. Through a human experimental study to test the immunomodulatory properties of metformin, an anti-diabetic drug, we chanced upon a functional determinant of neutralizing antibodies. Although vaccine viremia is a known correlate of antibody response, we found that in healthy volunteers with no detectable or low yellow fever 17D viremia, metformin-treated volunteers elicited higher neutralizing antibody titers than placebo-treated volunteers. Transcriptional and metabolomic analyses collectively showed that a brief course of metformin, started 3 days prior to YF17D vaccination and stopped at 3 days after vaccination, expanded oxidative phosphorylation and protein translation capacities. These increased capacities directly correlated with YF17D neutralizing antibody titers, with reduced reactive oxygen species response compared to placebo-treated volunteers. Our findings thus demonstrate a functional association between cellular respiration and vaccine-induced humoral immunity and suggest potential approaches to enhancing vaccine immunogenicity.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Metformina , Vacuna contra la Fiebre Amarilla , Humanos , Vacuna contra la Fiebre Amarilla/inmunología , Vacuna contra la Fiebre Amarilla/administración & dosificación , Metformina/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Inmunogenicidad Vacunal , Fiebre Amarilla/prevención & control , Fiebre Amarilla/inmunología , Adulto , Masculino , Femenino
3.
Genome Biol ; 24(1): 279, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38053173

RESUMEN

BACKGROUND: Identifying host factors is key to understanding RNA virus pathogenicity. Besides proteins, RNAs can interact with virus genomes to impact replication. RESULTS: Here, we use proximity ligation sequencing to identify virus-host RNA interactions for four strains of Zika virus (ZIKV) and one strain of dengue virus (DENV-1) in human cells. We find hundreds of coding and non-coding RNAs that bind to DENV and ZIKV viruses. Host RNAs tend to bind to single-stranded regions along the virus genomes according to hybridization energetics. Compared to SARS-CoV-2 interactors, ZIKV-interacting host RNAs tend to be downregulated upon virus infection. Knockdown of several short non-coding RNAs, including miR19a-3p, and 7SK RNA results in a decrease in viral replication, suggesting that they act as virus-permissive factors. In addition, the 3'UTR of DYNLT1 mRNA acts as a virus-restrictive factor by binding to the conserved dumbbell region on DENV and ZIKV 3'UTR to decrease virus replication. We also identify a conserved set of host RNAs that interacts with DENV, ZIKV, and SARS-CoV-2, suggesting that these RNAs are broadly important for RNA virus infection. CONCLUSIONS: This study demonstrates that host RNAs can impact virus replication in permissive and restrictive ways, expanding our understanding of host factors and RNA-based gene regulation during viral pathogenesis.


Asunto(s)
Virus del Dengue , Dengue , Infección por el Virus Zika , Virus Zika , Humanos , Virus Zika/genética , Infección por el Virus Zika/genética , ARN Viral/genética , Regiones no Traducidas 3' , Virus del Dengue/genética , Virus del Dengue/metabolismo , Replicación Viral , Dengue/genética , Antivirales , Dineínas/genética , Dineínas/metabolismo
4.
Open Biol ; 12(12): 220227, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36514984

RESUMEN

The four dengue viruses (DENVs) have evolved multiple mechanisms to ensure its survival. Among these mechanisms is the ability to regulate its replication rate, which may contribute to avoiding premature immune activation that limit infection dissemination: DENVs associated with dengue epidemics have shown slower replication rate than pre-epidemic strains. Correspondingly, wild-type DENVs replicate more slowly than their clinically attenuated derivatives. To understand how DENVs 'make haste slowly', we generated and screened for DENV2 mutants with accelerated replication that also induced high type-I interferon (IFN) expression in infected cells. We chanced upon a single NS2B-I114T amino acid substitution, in an otherwise highly conserved amino acid residue. Accelerated DENV2 replication damaged host DNA as mutant infection was dependent on host DNA damage repair factors, namely RAD21, EID3 and NEK5. DNA damage induced cGAS/STING signalling and activated early type-I IFN response that inhibited infection dissemination. Unexpectedly, STING activation also supported mutant DENV replication in infected cells through STING-induced autophagy. Our findings thus show that DENV NS2B has multi-faceted role in controlling DENV replication rate and immune evasion and suggest that the dual role of STING in supporting virus replication within infected cells but inhibiting infection dissemination could be particularly advantageous for live attenuated vaccine development.


Asunto(s)
Virus del Dengue , Interferón Tipo I , Evasión Inmune , Replicación Viral , Interferón Tipo I/genética , Transducción de Señal
5.
Cell Rep ; 31(6): 107617, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32402284

RESUMEN

The molecular basis of dengue virus (DENV) attenuation remains ambiguous and hampers a targeted approach to derive safe but nonetheless immunogenic live vaccine candidates. Here, we take advantage of DENV serotype 2 PDK53 vaccine strain, which recently and successfully completed a phase-3 clinical trial, to identify how this virus is attenuated compared to its wild-type parent, DENV2 16681. Site-directed mutagenesis on a 16681 infectious clone identifies a single G53D substitution in the non-structural 1 (NS1) protein that reduces 16681 infection and dissemination in both Aedes aegypti, as well as in mammalian cells to produce the characteristic phenotypes of PDK53. Mechanistically, NS1 G53D impairs the function of a known host factor, the endoplasmic reticulum (ER)-resident ribophorin 1 protein, to properly glycosylate NS1 and thus induce a host antiviral gene through ER stress responses. Our findings provide molecular insights on DENV attenuation on a clinically tested strain.


Asunto(s)
Vacunas contra el Dengue/farmacología , Virus del Dengue/genética , Virus del Dengue/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Aedes/virología , Animales , Chlorocebus aethiops , Dengue/virología , Vacunas contra el Dengue/inmunología , Estrés del Retículo Endoplásmico , Femenino , Glicosilación , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Mutagénesis Sitio-Dirigida , Mutación , Células Vero , Proteínas no Estructurales Virales/metabolismo
6.
Nat Commun ; 10(1): 1408, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926818

RESUMEN

Dengue (DENV) and Zika (ZIKV) viruses are clinically important members of the Flaviviridae family with an 11 kb positive strand RNA genome that folds to enable virus function. Here, we perform structure and interaction mapping on four DENV and ZIKV strains inside virions and in infected cells. Comparative analysis of SHAPE reactivities across serotypes nominates potentially functional regions that are highly structured, conserved, and contain low synonymous mutation rates. Interaction mapping by SPLASH identifies many pair-wise interactions, 40% of which form alternative structures, suggesting extensive structural heterogeneity. Analysis of shared interactions between serotypes reveals a conserved macro-organization whereby interactions can be preserved at physical locations beyond sequence identities. We further observe that longer-range interactions are preferentially disrupted inside cells, and show the importance of new interactions in virus fitness. These findings deepen our understanding of Flavivirus genome organization and serve as a resource for designing therapeutics in targeting RNA viruses.


Asunto(s)
Mapeo Cromosómico , Virus del Dengue/química , Virus del Dengue/genética , Virus Zika/química , Virus Zika/genética , Animales , Secuencia de Bases , Línea Celular , Secuencia Conservada , Genoma Viral , Humanos , Ratones , Modelos Moleculares , Mutación/genética , Ácidos Nicotínicos , ARN Viral/química , Virión/genética
7.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626665

RESUMEN

Airway epithelial cells and macrophages differ markedly in their responses to influenza A virus (IAV) infection. To investigate transcriptional responses underlying these differences, purified subsets of type II airway epithelial cells (ATII) and alveolar macrophages (AM) recovered from the lungs of mock- or IAV-infected mice at 9 h postinfection were subjected to RNA sequencing. This time point was chosen to allow for characterization of cell types first infected with the virus inoculum, prior to multicycle virus replication and the infiltration of inflammatory cells into the airways. In the absence of infection, AM predominantly expressed genes related to immunity, whereas ATII expressed genes consistent with their physiological roles in the lung. Following IAV infection, AM almost exclusively activated cell-intrinsic antiviral pathways that were dependent on interferon (IFN) regulatory factor 3/7 (IRF3/7) and/or type I IFN signaling. In contrast, IAV-infected ATII activated a broader range of physiological responses, including cell-intrinsic antiviral pathways, which were both independent of and dependent on IRF3/7 and/or type I IFN. These data suggest that transcriptional profiles hardwired during development are a major determinant underlying the different responses of ATII and AM to IAV infection.IMPORTANCE Airway epithelial cells (AEC) and airway macrophages (AM) represent major targets of influenza A virus (IAV) infection in the lung, yet the two cell types respond very differently to IAV infection. We have used RNA sequencing to define the host transcriptional responses in each cell type under steady-state conditions as well as following IAV infection. To do this, different cell subsets isolated from the lungs of mock- and IAV-infected mice were subjected to RNA sequencing. Under steady-state conditions, AM and AEC express distinct transcriptional activities, consistent with distinct physiological roles in the airways. Not surprisingly, these cells also exhibited major differences in transcriptional responses following IAV infection. These studies shed light on how the different transcriptional architectures of airway cells from two different lineages drive transcriptional responses to IAV infection.


Asunto(s)
Células Epiteliales/virología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Pulmón/virología , Macrófagos/virología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Animales , Línea Celular , Enfermedades Transmisibles/metabolismo , Enfermedades Transmisibles/virología , Perros , Células Epiteliales/metabolismo , Humanos , Gripe Humana/metabolismo , Gripe Humana/virología , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferones/metabolismo , Pulmón/metabolismo , Macrófagos/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Replicación Viral/fisiología
8.
Nat Commun ; 9(1): 1031, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29531213

RESUMEN

Zika virus (ZIKV) is a flavivirus that can cause congenital disease and requires development of an effective long-term preventative strategy. A replicative ZIKV vaccine with properties similar to the yellow fever 17D (YF17D) live-attenuated vaccine (LAV) would be advantageous, as a single dose of YF17D produces lifelong immunity. However, a replicative ZIKV vaccine must also be safe from causing persistent organ infections. Here we report an approach to ZIKV LAV development. We identify a ZIKV variant that produces small plaques due to interferon (IFN)-restricted viral propagation and displays attenuated infection of endothelial cells. We show that these properties collectively reduce the risk of organ infections and vertical transmission in a mouse model but remain sufficiently immunogenic to prevent wild-type ZIKV infection. Our findings suggest a strategy for the development of a safe but efficacious ZIKV LAV.


Asunto(s)
Técnicas Inmunológicas , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Infección por el Virus Zika/prevención & control , Virus Zika/genética , Virus Zika/inmunología , Aedes/inmunología , Aedes/virología , Animales , Células Dendríticas/inmunología , Células Dendríticas/virología , Humanos , Ratones , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Virus Zika/crecimiento & desarrollo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
9.
RNA ; 24(6): 803-814, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29572260

RESUMEN

The four dengue viruses (DENV1-4) are rapidly reemerging infectious RNA viruses. These positive-strand viral genomes contain structured 3' untranslated regions (UTRs) that interact with various host RNA binding proteins (RBPs). These RBPs are functionally important in viral replication, pathogenesis, and defense against host immune mechanisms. Here, we combined RNA chromatography and quantitative mass spectrometry to identify proteins interacting with DENV1-4 3' UTRs. As expected, RBPs displayed distinct binding specificity. Among them, we focused on quaking (QKI) because of its preference for the DENV4 3' UTR (DENV-4/SG/06K2270DK1/2005). RNA immunoprecipitation experiments demonstrated that QKI interacted with DENV4 genomes in infected cells. Moreover, QKI depletion enhanced infectious particle production of DENV4. On the contrary, QKI did not interact with DENV2 3' UTR, and DENV2 replication was not affected consistently by QKI depletion. Next, we mapped the QKI interaction site and identified a QKI response element (QRE) in DENV4 3' UTR. Interestingly, removal of QRE from DENV4 3' UTR abolished this interaction and increased DENV4 viral particle production. Introduction of the QRE to DENV2 3' UTR led to QKI binding and reduced DENV2 infectious particle production. Finally, reporter assays suggest that QKI reduced translation efficiency of viral RNA. Our work describes a novel function of QKI in restricting viral replication.


Asunto(s)
Regiones no Traducidas 3' , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Dengue/prevención & control , ARN Viral/genética , Proteínas de Unión al ARN/metabolismo , Replicación Viral/efectos de los fármacos , Dengue/genética , Dengue/virología , Genoma Viral , Células HEK293 , Humanos , Proteínas de Unión al ARN/genética
10.
Viruses ; 9(6)2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28587300

RESUMEN

Flaviviruses are enveloped arthropod-borne viruses with a single-stranded, positive-sense RNA genome that can cause serious illness in humans and animals. The 11 kb 5' capped RNA genome consists of a single open reading frame (ORF), and is flanked by 5' and 3' untranslated regions (UTR). The ORF is a polyprotein that is processed into three structural and seven non-structural proteins. The UTRs have been shown to be important for viral replication and immune modulation. Both of these regions consist of elements that are essential for genome cyclization, resulting in initiation of RNA synthesis. Genome mutation studies have been employed to investigate each component of the essential elements to show the necessity of each component and its role in viral RNA replication and growth. Furthermore, the highly structured 3'UTR is responsible for the generation of subgenomic flavivirus RNA (sfRNA) that helps the virus evade host immune response, thereby affecting viral pathogenesis. In addition, changes within the 3'UTR have been shown to affect transmissibility between vector and host, which can influence the development of vaccines.


Asunto(s)
Regiones no Traducidas 3' , Regiones no Traducidas 5' , Flavivirus/genética , Flavivirus/fisiología , Replicación Viral
11.
J Virol ; 91(14)2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28446666

RESUMEN

We previously identified a novel inhibitor of influenza virus in mouse saliva that halts the progression of susceptible viruses from the upper to the lower respiratory tract of mice in vivo and neutralizes viral infectivity in MDCK cells. Here, we investigated the viral target of the salivary inhibitor by using reverse genetics to create hybrid viruses with some surface proteins derived from an inhibitor-sensitive strain and others from an inhibitor-resistant strain. These viruses demonstrated that the origin of the viral neuraminidase (NA), but not the hemagglutinin or matrix protein, was the determinant of susceptibility to the inhibitor. Comparison of the NA sequences of a panel of H3N2 viruses with differing sensitivities to the salivary inhibitor revealed that surface residues 368 to 370 (N2 numbering) outside the active site played a key role in resistance. Resistant viruses contained an EDS motif at this location, and mutation to either EES or KDS, found in highly susceptible strains, significantly increased in vitro susceptibility to the inhibitor and reduced the ability of the virus to progress to the lungs when the viral inoculum was initially confined to the upper respiratory tract. In the presence of saliva, viral strains with a susceptible NA could not be efficiently released from the surfaces of infected MDCK cells and had reduced enzymatic activity based on their ability to cleave substrate in vitro This work indicates that the mouse has evolved an innate inhibitor similar in function, though not in mechanism, to what humans have created synthetically as an antiviral drug for influenza virus.IMPORTANCE Despite widespread use of experimental pulmonary infection of the laboratory mouse to study influenza virus infection and pathogenesis, to our knowledge, mice do not naturally succumb to influenza. Here, we show that mice produce their own natural form of neuraminidase inhibitor in saliva that stops the virus from reaching the lungs, providing a possible mechanism through which the species may not experience severe influenza virus infection in the wild. We show that the murine salivary inhibitor targets the outer surface of the influenza virus neuraminidase, possibly occluding entry to the enzymatic site rather than binding within the active site like commercially available neuraminidase inhibitors. This knowledge sheds light on how the natural inhibitors of particular species combat infection.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Pulmón/virología , Neuraminidasa/antagonistas & inhibidores , Sistema Respiratorio/virología , Saliva/inmunología , Proteínas Virales/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Perros , Inmunidad Innata , Subtipo H3N2 del Virus de la Influenza A/genética , Células de Riñón Canino Madin Darby , Ratones , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Genética Inversa , Proteínas Virales/genética
12.
Sci Rep ; 6: 19428, 2016 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-26763587

RESUMEN

The ubiquitous presence of cell-surface sialic acid (SIA) has complicated efforts to identify specific transmembrane glycoproteins that function as bone fide entry receptors for influenza A virus (IAV) infection. The C-type lectin receptors (CLRs) DC-SIGN (CD209) and L-SIGN (CD209L) enhance IAV infection however it is not known if they act as attachment factors, passing virions to other unknown receptors for virus entry, or as authentic entry receptors for CLR-mediated virus uptake and infection. Sialic acid-deficient Lec2 Chinese Hamster Ovary (CHO) cell lines were resistant to IAV infection whereas expression of DC-SIGN/L-SIGN restored susceptibility of Lec2 cells to pH- and dynamin-dependent infection. Moreover, Lec2 cells expressing endocytosis-defective DC-SIGN/L-SIGN retained capacity to bind IAV but showed reduced susceptibility to infection. These studies confirm that DC-SIGN and L-SIGN are authentic endocytic receptors for IAV entry and infection.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Endocitosis , Virus de la Influenza A/fisiología , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Internalización del Virus , Animales , Células CHO , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Línea Celular , Cricetulus , Perros , Dinaminas/metabolismo , Expresión Génica , Concentración de Iones de Hidrógeno , Lectinas Tipo C/química , Lectinas Tipo C/genética , Mutación , Ácido N-Acetilneuramínico/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Acoplamiento Viral
13.
J Virol ; 90(1): 206-21, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26468543

RESUMEN

UNLABELLED: It is well established that influenza A virus (IAV) attachment to and infection of epithelial cells is dependent on sialic acid (SIA) at the cell surface, although the specific receptors that mediate IAV entry have not been defined and multiple receptors may exist. Lec2 Chinese hamster ovary (CHO) cells are SIA deficient and resistant to IAV infection. Here we demonstrate that the expression of the C-type lectin receptor langerin in Lec2 cells (Lec2-Lg) rendered them permissive to IAV infection, as measured by replication of the viral genome, transcription of viral mRNA, and synthesis of viral proteins. Unlike SIA-dependent infection of parental CHO cells, IAV attachment and infection of Lec2-Lg cells was mediated via lectin-mediated recognition of mannose-rich glycans expressed by the viral hemagglutinin glycoprotein. Lec2 cells expressing endocytosis-defective langerin bound IAV efficiently but remained resistant to IAV infection, confirming that internalization via langerin was essential for infectious entry. Langerin-mediated infection of Lec2-Lg cells was pH and dynamin dependent, occurred via clathrin- and caveolin-mediated endocytic pathways, and utilized early (Rab5(+)) but not late (Rab7(+)) endosomes. This study is the first to demonstrate that langerin represents an authentic receptor that binds and internalizes IAV to facilitate infection. Moreover, it describes a unique experimental system to probe specific pathways and compartments involved in infectious entry following recognition of IAV by a single cell surface receptor. IMPORTANCE: On the surface of host cells, sialic acid (SIA) functions as the major attachment factor for influenza A viruses (IAV). However, few studies have identified specific transmembrane receptors that bind and internalize IAV to facilitate infection. Here we identify human langerin as a transmembrane glycoprotein that can act as an attachment factor and a bone fide endocytic receptor for IAV infection. Expression of langerin by an SIA-deficient cell line resistant to IAV rendered cells permissive to infection. As langerin represented the sole receptor for IAV infection in this system, we have defined the pathways and compartments involved in infectious entry of IAV into cells following recognition by langerin.


Asunto(s)
Antígenos CD/metabolismo , Virus de la Influenza A/fisiología , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Receptores Virales/metabolismo , Acoplamiento Viral , Internalización del Virus , Animales , Células CHO , Cricetulus , Dinaminas/metabolismo , Endocitosis , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Polisacáridos/metabolismo , Unión Proteica
14.
PLoS One ; 10(11): e0143539, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26600246

RESUMEN

Respiratory dendritic cells (DC) play a pivotal role in the initiation of adaptive immune responses to influenza virus. To do this, respiratory DCs must ferry viral antigen from the lung to the draining lymph node without becoming infected and perishing en route. We show that respiratory DCs up-regulate the expression of the antiviral molecule, interferon-induced transmembrane protein 3 (IFITM3) in response to influenza virus infection, in a manner dependent on type I interferon signaling and the transcription factors IRF7 and IRF3. Failure of respiratory DCs to up-regulate IFITM3 following influenza virus infection resulted in impaired trafficking to the draining LN and consequently in impaired priming of an influenza-specific CD8+ T cell response. The impaired trafficking of IFITM3-deficient DC correlated with an increased susceptibility of these DC to influenza virus infection. This work shows that the expression of IFITM3 protects respiratory DCs from influenza virus infection, permitting migration from lung to LN and optimal priming of a virus specific T-cell response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Animales , Western Blotting , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Citometría de Flujo , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/metabolismo
15.
J Antimicrob Chemother ; 69(8): 2164-74, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24777908

RESUMEN

OBJECTIVES: Emerging drug resistance to antiviral therapies is an increasing challenge for the treatment of influenza virus infections. One new antiviral compound, BTA938, a dimeric derivative of the viral neuraminidase inhibitor zanamivir, contains a 14-carbon linker bridging two zanamivir moieties. In these studies, we evaluated antiviral efficacy in cell cultures infected with influenza virus and in mouse models of lethal influenza using H1N1pdm09, H3N2 and oseltamivir-resistant (H275Y) viruses. METHODS: In vitro activity was evaluated against 22 strains of influenza virus. Additionally, in vivo studies compared the efficacy of BTA938 or zanamivir after intranasal treatment. We also tested the hypothesis of a dual mode of action for BTA938 using scanning electron microscopy (SEM). RESULTS: BTA938 inhibited the viruses at nanomolar concentrations in vitro with a median 50% effective concentration value of 0.5 nM. In mouse models, the dimer provided ∼10-fold greater protection than zanamivir. The data also showed that a single low dose (3 mg/kg) protected 100% of mice from an otherwise lethal oseltamivir-resistant (H275Y) influenza virus infection. Remarkably, a single prophylactic treatment (10 mg/kg) administered 7 days before the challenge protected 70% of mice and when administered 1 or 3 days before the challenge it protected 90% of mice. Additionally, SEM provides evidence that the increased antiviral potency may be mediated by an enhanced aggregation of virus on the cell surface. CONCLUSIONS: In vitro and in vivo experiments showed the high antiviral activity of BTA938 for the treatment of influenza virus infections. Moreover, we demonstrated that a single dose of BTA938 is sufficient for prophylactic and therapeutic protection in mouse models.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Zanamivir/análogos & derivados , Zanamivir/farmacología , Animales , Antivirales/farmacología , Perros , Combinación de Medicamentos , Farmacorresistencia Viral , Inhibidores Enzimáticos/farmacología , Femenino , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/clasificación , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/virología , Oseltamivir/farmacología
16.
J Virol ; 88(3): 1659-72, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24257596

RESUMEN

Specific protein receptors that mediate internalization and entry of influenza A virus (IAV) have not been identified for any cell type. Sialic acid (SIA), the primary attachment factor for IAV hemagglutinin, is expressed by numerous cell surface glycoproteins and glycolipids, confounding efforts to identify specific receptors involved in virus infection. Lec1 Chinese hamster ovary (CHO) epithelial cells express cell surface SIA and bind IAV yet are largely resistant to infection. Here, we demonstrate that expression of the murine macrophage galactose-type lectin 1 (MGL1) by Lec1 cells enhanced Ca(2+)-dependent IAV binding and restored permissivity to infection. Lec1 cells expressing MGL1 were infected in the presence or absence of cell surface SIA, indicating that MGL1 can act as a primary receptor or as a coreceptor with SIA. Lec1 cells expressing endocytosis-deficient MGL1 mediated Ca(2+)-dependent IAV binding but were less sensitive to IAV infection, indicating that direct internalization via MGL1 can result in cellular infection. Together, these studies identify MGL1 as a cell surface glycoprotein that can act as an authentic receptor for both attachment and infectious entry of IAV.


Asunto(s)
Asialoglicoproteínas/metabolismo , Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Virales/metabolismo , Acoplamiento Viral , Internalización del Virus , Animales , Asialoglicoproteínas/genética , Células CHO , Calcio/metabolismo , Línea Celular , Cricetinae , Cricetulus , Humanos , Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/virología , Lectinas Tipo C/genética , Macrófagos/metabolismo , Macrófagos/virología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Receptores Virales/genética
17.
J Leukoc Biol ; 93(1): 145-54, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23108101

RESUMEN

The helper-dependent pathway of priming CD8(+) T cells involves "licensing" of DCs by CD40L on CD4(+) T cells. The helper-independent ("helpless") pathways elicited by many viruses, including influenza, are less widely understood. We have postulated that CD40L can be up-regulated on DCs by such viruses, and this promotes priming of CD8(+) T cells via CD40. Most studies on costimulation have been performed in the presence of CD4(+) T cells, and so the role of CD40L costimulation under helpless circumstances has not been fully elucidated. Here, we investigated such a role for CD40L using CD40L KO mice. Although the number of influenza-specific CD8(+) T cells was unaffected by the absence of CD4(+) T cells, it was markedly decreased in the absence of CD40L. Proliferation (the number of CD44(+)BrdU(+) influenza-specific CD8(+) T cells) in the primary response was diminished in CD40L KO mice at Day 8 but not at Day 5 after infection. MLR studies indicated that CD40L expression on DCs was critical for CD8(+) T cell activation. Adoptive transfer of CD40 KO CD8(+) T cells compared with WT cells confirmed that CD40 on such cells was critical for the generation of primary anti-influenza CD8(+) T cell responses. The late effect also corresponded with the late expression of CD40 by influenza-specific CD8(+) T cells. We suggest that costimulation via CD40L on DCs and CD40 on CD8(+) T cells is important in optimizing primary CD8(+) T cell responses during influenza infection.


Asunto(s)
Antígenos CD40/inmunología , Ligando de CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/metabolismo , Ensayo de Inmunoadsorción Enzimática , Virus de la Influenza A , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/metabolismo
18.
J Biomed Biotechnol ; 2012: 732191, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22665991

RESUMEN

Host defenses against viral infections depend on a complex interplay of innate (nonspecific) and adaptive (specific) components. In the early stages of infection, innate mechanisms represent the main line of host defense, acting to limit the spread of virus in host tissues prior to the induction of the adaptive immune response. Serum and lung fluids contain a range of lectins capable of recognizing and destroying influenza A viruses (IAV). Herein, we review the mechanisms by which soluble endogenous lectins mediate anti-IAV activity, including their role in modulating IAV-induced inflammation and disease and their potential as prophylactic and/or therapeutic treatments during severe IAV-induced disease.


Asunto(s)
Virus de la Influenza A/inmunología , Lectinas/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Animales , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata
19.
PLoS One ; 5(10): e13622, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-21049034

RESUMEN

BACKGROUND: Despite the availability of specific vaccines and antiviral drugs, influenza continues to impose a heavy toll on human health worldwide. Passive transfer of specific antibody (Ab) may provide a useful means of preventing or treating disease in unvaccinated individuals or those failing to adequately seroconvert, especially now that resistance to antiviral drugs is on the rise. However, preparation of appropriate Ab in large scale, quickly and on a yearly basis is viewed as a significant logistical hurdle for this approach to control seasonal influenza. METHODOLOGY/PRINCIPAL FINDINGS: In this study, bovine colostrum, which contains approximately 500 g of IgG per milking per animal, has been investigated as a source of polyclonal antibody for delivery to the respiratory tract. IgG and F(ab')2 were purified from the hyperimmune colostrum of cows vaccinated with influenza A/Puerto Rico/8/34 (PR8) vaccine and were shown to have high hemagglutination-inhibitory and virus-neutralizing titers. In BALB/c mice, a single administration of either IgG or F(ab')2 could prevent the establishment of infection with a sublethal dose of PR8 virus when given as early as 7 days prior to exposure to virus. Pre-treated mice also survived an otherwise lethal dose of virus, the IgG- but not the F(ab')2-treated mice showing no weight loss. Successful reduction of established infection with this highly virulent virus was also observed with a single treatment 24 hr after virus exposure. CONCLUSIONS/SIGNIFICANCE: These data suggest that a novel and commercially-scalable technique for preparing Ab from hyperimmune bovine colostrum could allow production of a valuable substitute for antiviral drugs to control influenza with the advantage of eliminating the need for daily administration.


Asunto(s)
Anticuerpos/uso terapéutico , Calostro/inmunología , Infecciones por Orthomyxoviridae/terapia , Animales , Bovinos , Electroforesis en Gel de Poliacrilamida , Inmunoglobulinas/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización
20.
Influenza Other Respir Viruses ; 3(4): 177-82, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19627375

RESUMEN

BACKGROUND: The best form of protection against influenza is high-titred virus-neutralizing antibody specific for the challenge strain. However, this is not always possible to achieve by vaccination due to the need for predicting the emerging virus, whether it be a drift variant of existing human endemic influenza type A subtypes or the next pandemic virus, for incorporation into the vaccine. By activating additional arms of the immune system to provide heterosubtypic immunity, that is immunity active against all viruses of type A influenza regardless of subtype or strain, it should be possible to provide significant benefit in situations where appropriate antibody responses are not achieved. Although current inactivated vaccines are unable to induce heterosubtypic CD8(+) T cell immunity, we have shown that lipopeptides are particularly efficient in this regard. OBJECTIVES: To examine the role of vaccine-induced CD8(+) T cells in altering the course of disease due to highly virulent H1N1 influenza virus in the mouse model. METHODS: The induction of influenza-specific CD8(+) T cells following intranasal inoculation with lipopeptide vaccine was assessed by intracellular cytokine staining (ICS) and the capacity of these cells to reduce viral loads in the lungs and to protect against death after viral challenge was determined. RESULTS AND CONCLUSIONS: We show that CD8(+) T cells are induced by a single intranasal vaccination with lipopeptide, they remain at substantial levels in the lungs and are efficiently boosted upon challenge with virulent virus to provide late control of pulmonary viral loads. Vaccinated mice are not only protected from death but remain active, indicative of less severe disease despite significant weight loss.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Lipopéptidos/inmunología , Administración Intranasal , Animales , Peso Corporal , Citocinas/biosíntesis , Vacunas contra la Influenza/administración & dosificación , Lipopéptidos/administración & dosificación , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...