Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Ther ; 30(5): 2024-2047, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34999208

RESUMEN

Conventional influenza vaccines fail to confer broad protection against diverse influenza A viruses with pandemic potential. Efforts to develop a universal influenza virus vaccine include refocusing immunity towards the highly conserved stalk domain of the influenza virus surface glycoprotein, hemagglutinin (HA). We constructed a non-replicating adenoviral (Ad) vector, encoding a secreted form of H1 HA, to evaluate HA stalk-focused immunity. The Ad5_H1 vaccine was tested in mice for its ability to elicit broad, cross-reactive protection against homologous, heterologous, and heterosubtypic lethal challenge in a single-shot immunization regimen. Ad5_H1 elicited hemagglutination inhibition (HI+) active antibodies (Abs), which conferred 100% sterilizing protection from homologous H1N1 challenge. Furthermore, Ad5_H1 rapidly induced H1-stalk-specific Abs with Fc-mediated effector function activity, in addition to stimulating both CD4+ and CD8+ stalk-specific T cell responses. This phenotype of immunity provided 100% protection from lethal challenge with a head-mismatched, reassortant influenza virus bearing a chimeric HA, cH6/1, in a stalk-mediated manner. Most importantly, 100% protection from mortality following lethal challenge with a heterosubtypic avian influenza virus, H5N1, was observed following a single immunization with Ad5_H1. In conclusion, Ad-based influenza vaccines can elicit significant breadth of protection in naive animals and could be considered for pandemic preparedness and stockpiling.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Adenoviridae/genética , Animales , Anticuerpos Antivirales , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Hemaglutininas , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Humana/prevención & control , Ratones , Ratones Endogámicos BALB C
2.
Antimicrob Agents Chemother ; 59(7): 4162-72, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25941218

RESUMEN

The emergence of influenza virus strains resistant to approved neuraminidase inhibitors and the time constrains after infection when these drugs can be effective constitute major drawbacks for this class of drugs. This highlights a critical need to discover new therapeutic agents that can be used for the treatment of influenza virus-infected patients. The use of broadly neutralizing anti-influenza monoclonal antibodies (MAbs) has been sought as an alternative immunotherapy against influenza infection. Here, we tested in mice previously characterized broadly neutralizing anti-hemagglutinin (HA) stalk MAbs prophylactically and therapeutically using different routes of administration. The efficacy of treatment against an influenza H1N1 pandemic virus challenge was compared between two systemic routes of administration, intraperitoneal (i.p.) and intravenous (i.v.), and two local routes, intranasal (i.n.) and aerosol (a.e.). The dose of MAb required for prophylactic protection was reduced by 10-fold in animals treated locally (i.n. or a.e.) compared with those treated systemically (i.p. or i.v.). Improved therapeutic protection was observed in animals treated i.n. on day 5 postinfection (60% survival) compared with those treated via the i.p. route (20% survival). An increase in therapeutic efficacy against other influenza virus subtypes (H5N1) was also observed when a local route of administration was used. Our findings demonstrate that local administration significantly decreases the amount of broadly neutralizing monoclonal antibody required for protection against influenza, which highlights the potential use of MAbs as a therapeutic agent for influenza-associated disease.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/uso terapéutico , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Gripe Humana/tratamiento farmacológico , Administración Intranasal , Administración Intravenosa , Aerosoles , Animales , Anticuerpos Monoclonales/farmacocinética , Antivirales/farmacocinética , Disponibilidad Biológica , Femenino , Hemaglutininas/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/patología , Gripe Humana/virología , Inyecciones Intraperitoneales , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Distribución Tisular
3.
J Virol ; 88(6): 3432-42, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24403585

RESUMEN

UNLABELLED: Therapeutic monoclonal antibodies that target the conserved stalk domain of the influenza virus hemagglutinin and stalk-based universal influenza virus vaccine strategies are being developed as promising countermeasures for influenza virus infections. The pan-H1-reactive monoclonal antibody 6F12 has been extensively characterized and shows broad efficacy against divergent H1N1 strains in the mouse model. Here we demonstrate its efficacy against a pandemic H1N1 challenge virus in the ferret model of influenza disease. Furthermore, we recently developed a universal influenza virus vaccine strategy based on chimeric hemagglutinin constructs that focuses the immune response on the conserved stalk domain of the hemagglutinin. Here we set out to test this vaccination strategy in the ferret model. Both strategies, pretreatment of animals with a stalk-reactive monoclonal antibody and vaccination with chimeric hemagglutinin-based constructs, were able to significantly reduce viral titers in nasal turbinates, lungs, and olfactory bulbs. In addition, vaccinated animals also showed reduced nasal wash viral titers. In summary, both strategies showed efficacy in reducing viral loads after an influenza virus challenge in the ferret model. IMPORTANCE: Influenza virus hemagglutinin stalk-reactive antibodies tend to be less potent yet are more broadly reactive and can neutralize seasonal and pandemic influenza virus strains. The ferret model was used to assess the potential of hemagglutinin stalk-based immunity to provide protection against influenza virus infection. The novelty and significance of the findings described in this report support the development of vaccines stimulating stalk-specific antibody responses.


Asunto(s)
Modelos Animales de Enfermedad , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Hemaglutininas , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/química , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Masculino , Estructura Terciaria de Proteína
4.
J Virol ; 87(15): 8272-81, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23698304

RESUMEN

Influenza A virus is a major human pathogen responsible for seasonal epidemics as well as pandemic outbreaks. Due to the continuing burden on human health, the need for new tools to study influenza virus pathogenesis as well as to evaluate new therapeutics is paramount. We report the development of a stable, replication-competent luciferase reporter influenza A virus that can be used for in vivo imaging of viral replication. This imaging is noninvasive and allows for the longitudinal monitoring of infection in living animals. We used this tool to characterize novel monoclonal antibodies that bind the conserved stalk domain of the viral hemagglutinin of H1 and H5 subtypes and protect mice from lethal disease. The use of luciferase reporter influenza viruses allows for new mechanistic studies to expand our knowledge of virus-induced disease and provides a new quantitative method to evaluate future antiviral therapies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/farmacología , Virus de la Influenza A/patogenicidad , Mediciones Luminiscentes/métodos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Imagen de Cuerpo Entero/métodos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Antivirales/administración & dosificación , Modelos Animales de Enfermedad , Genes Reporteros , Virus de la Influenza A/genética , Luciferasas/análisis , Luciferasas/genética , Ratones , Ratones Endogámicos BALB C , Coloración y Etiquetado/métodos , Análisis de Supervivencia , Resultado del Tratamiento
5.
PLoS Pathog ; 8(4): e1002668, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22577360

RESUMEN

Influenza viruses continue to pose a major public health threat worldwide and options for antiviral therapy are limited by the emergence of drug-resistant virus strains. The antiviral cytokine, interferon (IFN) is an essential mediator of the innate immune response and influenza viruses, like many viruses, have evolved strategies to evade this response, resulting in increased replication and enhanced pathogenicity. A cell-based assay that monitors IFN production was developed and applied in a high-throughput compound screen to identify molecules that restore the IFN response to influenza virus infected cells. We report the identification of compound ASN2, which induces IFN only in the presence of influenza virus infection. ASN2 preferentially inhibits the growth of influenza A viruses, including the 1918 H1N1, 1968 H3N2 and 2009 H1N1 pandemic strains and avian H5N1 virus. In vivo, ASN2 partially protects mice challenged with a lethal dose of influenza A virus. Surprisingly, we found that the antiviral activity of ASN2 is not dependent on IFN production and signaling. Rather, its IFN-inducing property appears to be an indirect effect resulting from ASN2-mediated inhibition of viral polymerase function, and subsequent loss of the expression of the viral IFN antagonist, NS1. Moreover, we identified a single amino acid mutation at position 499 of the influenza virus PB1 protein that confers resistance to ASN2, suggesting that PB1 is the direct target. This two-pronged antiviral mechanism, consisting of direct inhibition of virus replication and simultaneous activation of the host innate immune response, is a unique property not previously described for any single antiviral molecule.


Asunto(s)
Antivirales/farmacología , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Virus de la Influenza A/efectos de los fármacos , Interferones/biosíntesis , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Antivirales/química , Células Cultivadas , Perros , Inhibidores Enzimáticos/química , Haplorrinos , Humanos , Indoles/química , Virus de la Influenza A/fisiología , Ratones , Ratones Endogámicos BALB C , Replicación Viral/efectos de los fármacos
6.
Proc Natl Acad Sci U S A ; 108(37): 15366-71, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21896751

RESUMEN

Influenza nucleoprotein (NP) plays multiple roles in the virus life cycle, including an essential function in viral replication as an integral component of the ribonucleoprotein complex, associating with viral RNA and polymerase within the viral core. The multifunctional nature of NP makes it an attractive target for antiviral intervention, and inhibitors targeting this protein have recently been reported. In a parallel effort, we discovered a structurally similar series of influenza replication inhibitors and show that they interfere with NP-dependent processes via formation of higher-order NP oligomers. Support for this unique mechanism is provided by site-directed mutagenesis studies, biophysical characterization of the oligomeric ligand:NP complex, and an X-ray cocrystal structure of an NP dimer of trimers (or hexamer) comprising three NP_A:NP_B dimeric subunits. Each NP_A:NP_B dimeric subunit contains two ligands that bridge two composite, protein-spanning binding sites in an antiparallel orientation to form a stable quaternary complex. Optimization of the initial screening hit produced an analog that protects mice from influenza-induced weight loss and mortality by reducing viral titers to undetectable levels throughout the course of treatment.


Asunto(s)
Antivirales/farmacología , Nucleoproteínas/química , Nucleoproteínas/metabolismo , Orthomyxoviridae/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Ensayos Analíticos de Alto Rendimiento , Hidrodinámica , Ratones , Modelos Moleculares , Nucleoproteínas/ultraestructura , Orthomyxoviridae/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Multimerización de Proteína/efectos de los fármacos , Estructura Cuaternaria de Proteína , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Soluciones
7.
ACS Appl Bio Mater ; 7(6): 4133-4141, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38812435

RESUMEN

The ultimate vaccine against infections caused by Nipah virus should be capable of providing protection at the respiratory tract─the most probable port of entry for this pathogen. Intranasally delivered vaccines, which target nasal-associated lymphoid tissue and induce both systemic and mucosal immunity, are attractive candidates for enabling effective vaccination against this lethal disease. Herein, the water-soluble polyphosphazene delivery vehicle assembles into nanoscale supramolecular constructs with the soluble extracellular portion of the Hendra virus attachment glycoprotein─a promising subunit vaccine antigen against both Nipah and Hendra viruses. These supramolecular constructs signal through Toll-like receptor 7/8 and promote binding interactions with mucin─an important feature of effective mucosal adjuvants. High mass contrast of phosphorus-nitrogen backbone of the polymer enables a successful visualization of nanoconstructs in their vitrified state by cryogenic electron microscopy. Here, we characterize the self-assembly of polyphosphazene macromolecule with biologically relevant ligands by asymmetric flow field flow fractionation, dynamic light scattering, fluorescence spectrophotometry, and turbidimetric titration methods. Furthermore, a polyphosphazene-enabled intranasal Nipah vaccine candidate demonstrates the ability to induce immune responses in hamsters and shows superiority in inducing total IgG and neutralizing antibodies when benchmarked against the respective clinical stage alum adjuvanted vaccine. The results highlight the potential of polyphosphazene-enabled nanoassemblies in the development of intranasal vaccines.


Asunto(s)
Administración Intranasal , Virus Nipah , Compuestos Organofosforados , Polímeros , Vacunas de Subunidad , Vacunas Virales , Compuestos Organofosforados/química , Compuestos Organofosforados/administración & dosificación , Polímeros/química , Virus Nipah/inmunología , Animales , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/química , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/química , Vacunas de Subunidad/administración & dosificación , Tamaño de la Partícula , Ensayo de Materiales , Materiales Biocompatibles/química , Nanopartículas/química , Inmunización
8.
Emerg Infect Dis ; 18(7): 1135-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22710350

RESUMEN

To determine whether guinea pigs are infected with influenza virus in nature, we conducted a serologic study in domestic guinea pigs in Ecuador. Detection of antibodies against influenza A and B raises the question about the role of guinea pigs in the ecology and epidemiology of influenza virus in the region.


Asunto(s)
Anticuerpos Antivirales/sangre , Cobayas , Ganado , Infecciones por Orthomyxoviridae/veterinaria , Orthomyxoviridae/inmunología , Animales , Ecuador/epidemiología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Infecciones por Orthomyxoviridae/epidemiología , Infecciones por Orthomyxoviridae/virología
9.
Brain Behav Immun ; 24(2): 281-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19836444

RESUMEN

Mouse-adapted human influenza virus is detectable in the olfactory bulbs of mice within hours after intranasal challenge and is associated with enhanced local cytokine mRNA and protein levels. To determine whether signals from the olfactory nerve influence the unfolding of the acute phase response (APR), we surgically transected the olfactory nerve in mice prior to influenza infection. We then compared the responses of olfactory-nerve-transected (ONT) mice to those recorded in sham-operated control mice using measurements of body temperature, food intake, body weight, locomotor activity and immunohistochemistry for cytokines and the viral antigen, H1N1. ONT did not change baseline body temperature (Tb); however, the onset of virus-induced hypothermia was delayed for about 13 h in the ONT mice. Locomotor activity, food intake and body weights of the two groups were similar. At 15 h post-challenge fewer viral antigen-immunoreactive (IR) cells were observed in the olfactory bulb (OB) of ONT mice compared to sham controls. The number of tumor necrosis factor alpha (TNFalpha)- and interleukin 1beta (IL1beta)-IR cells in ONT mice was also reduced in the OB and other interconnected regions in the brain compared to sham controls. These results suggest that the olfactory nerve pathway is important for the initial pathogenesis of the influenza-induced APR.


Asunto(s)
Temperatura Corporal/fisiología , Química Encefálica/fisiología , Citocinas/biosíntesis , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/metabolismo , Gripe Humana/fisiopatología , Nervio Olfatorio/fisiopatología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/fisiopatología , Animales , Antígenos Virales/análisis , Peso Corporal/fisiología , Encéfalo/patología , Ingestión de Alimentos , Humanos , Inmunohistoquímica , Gripe Humana/psicología , Gripe Humana/virología , Interleucina-1beta/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Bulbo Olfatorio/patología , Nervio Olfatorio/patología , Infecciones por Orthomyxoviridae/psicología , Factor de Necrosis Tumoral alfa/biosíntesis
10.
Front Microbiol ; 10: 1327, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31275265

RESUMEN

Emergence and re-emergence of respiratory virus infections represent a significant threat to global public health, as they occur seasonally and less frequently (such as in the case of influenza virus) as pandemic infections. Some of these viruses have been in the human population for centuries and others had recently emerged as a public health problem. Influenza viruses have been affecting the human population for a long time now; however, their ability to rapidly evolve through antigenic drift and antigenic shift causes the emergence of new strains. A recent example of these events is the avian-origin H7N9 influenza virus outbreak currently undergoing in China. Human H7N9 influenza viruses are resistant to amantadines and some strains are also resistant to neuraminidase inhibitors greatly limiting the options for treatment. Respiratory syncytial virus (RSV) may cause a lower respiratory tract infection characterized by bronchiolitis and pneumonia mainly in children and the elderly. Infection with RSV can cause severe disease and even death, imposing a severe burden for pediatric and geriatric health systems worldwide. Treatment for RSV is mainly supportive since the only approved therapy, a monoclonal antibody, is recommended for prophylactic use in high-risk patients. The Middle East respiratory syndrome coronavirus (MERS-CoV) is a newly emerging respiratory virus. The virus was first recognized in 2012 and it is associated with a lower respiratory tract disease that is more severe in patients with comorbidities. No licensed vaccines or antivirals have been yet approved for the treatment of MERS-CoV in humans. It is clear that the discovery and development of novel antivirals that can be used alone or in combination with existing therapies to treat these important respiratory viral infections are critical. In this review, we will describe some of the novel therapeutics currently under development for the treatment of these infections.

11.
ACS Infect Dis ; 5(10): 1779-1793, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31448902

RESUMEN

Two classes of antivirals targeting the viral neuraminidase (NA) and endonuclease are currently the only clinically useful drugs for the treatment of influenza. However, resistance to both antivirals has been observed in clinical isolates, and there was widespread resistance to oseltamivir (an NA inhibitor) among H1N1 viruses prior to 2009. This potential for resistance and lack of diversity for antiviral targets highlights the need for new influenza antivirals with a higher barrier to resistance. In this study, we identified an antiviral compound, M85, that targets host kinases, epidermal growth factor receptor (EGFR), and phosphoinositide 3 class II ß (PIK3C2ß) and is not susceptible to resistance by viral mutations. M85 blocks endocytosis of influenza viruses and inhibits a broad-spectrum of viruses with minimal cytotoxicity. In vitro, we found that combinations of M85 and oseltamivir have strong synergism. In the mouse model for influenza, treatment with the combination therapy was more protective against a lethal viral challenge than oseltamivir alone, indicating that development of M85 could lead to combination therapies for influenza. Finally, through this discovery of M85 and its antiviral mechanism, we present the first description of PIK3C2ß as a necessary host factor for influenza virus entry.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Orthomyxoviridae/efectos de los fármacos , Oseltamivir/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Fosfatidilinositol 3-Quinasas Clase II/efectos de los fármacos , Modelos Animales de Enfermedad , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Farmacorresistencia Viral/efectos de los fármacos , Sinergismo Farmacológico , Receptores ErbB , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Células Vero
12.
Nat Commun ; 9(1): 4560, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30385750

RESUMEN

Zika virus is a mosquito-borne flavivirus closely related to dengue virus that can cause severe disease in humans, including microcephaly in newborns and Guillain-Barré syndrome in adults. Specific treatments and vaccines for Zika virus are not currently available. Here, we isolate and characterize four monoclonal antibodies (mAbs) from an infected patient that target the non-structural protein NS1. We show that while these antibodies are non-neutralizing, NS1-specific mAbs can engage FcγR without inducing antibody dependent enhancement (ADE) of infection in vitro. Moreover, we demonstrate that mAb AA12 has protective efficacy against lethal challenges of African and Asian lineage strains of Zika virus in Stat2-/- mice. Protection is Fc-dependent, as a mutated antibody unable to activate known Fc effector functions or complement is not protective in vivo. This study highlights the importance of the ZIKV NS1 protein as a potential vaccine antigen.


Asunto(s)
Anticuerpos Antivirales/metabolismo , Receptores de IgG/metabolismo , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/inmunología , Infección por el Virus Zika/prevención & control , Virus Zika/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/farmacología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Células Jurkat , Ratones , Ratones Noqueados , Pruebas de Neutralización , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT2/genética , Células Vero , Proteínas no Estructurales Virales/metabolismo , Virus Zika/metabolismo
13.
Antiviral Res ; 142: 12-15, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28286235

RESUMEN

Oseltamivir is an influenza neuraminidase inhibitor that along with supportive therapy has shown to help critically ill patients infected with H7N9 and H1N1pdm influenza virus strains to recover from disease. The standard of care recommends the administration of oseltamivir via oral route which represents difficulties in patients with gastrointestinal complications. Here we tested the use of aerosol administration of oseltamivir to treat mice infected with influenza A/H7N9 virus or influenza A/H1N1pdm virus and directly compared this approach to the standard of care, oral administration. Using nose only delivery of aerosolized oseltamivir we observed a significant increase in efficacy of the treatment compared to oral administration characterized by reduced body weight loss, increased survival rate and dose sparing. The preclinical data presented here supports the possibility of using this approach in clinical settings.


Asunto(s)
Aerosoles/administración & dosificación , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Oseltamivir/administración & dosificación , Oseltamivir/farmacología , Administración Oral , Animales , Antivirales/administración & dosificación , Antivirales/farmacología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Tasa de Supervivencia , Pérdida de Peso
14.
mBio ; 8(2)2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28351919

RESUMEN

An exacerbated immune response is one of the main causes of influenza-induced lung damage during infection. The molecular mechanisms regulating the fate of the initial immune response to infection, either as a protective response or as detrimental immunopathology, are not well understood. The purinergic receptor P2X7 is an ionotropic nucleotide-gated ion channel receptor expressed on immune cells that has been implicated in induction and maintenance of excessive inflammation. Here, we analyze the role of this receptor in a mouse model of influenza virus infection using a receptor knockout (KO) mouse strain. Our results demonstrate that the absence of the P2X7 receptor results in a better outcome to influenza virus infection characterized by reduced weight loss and increased survival upon experimental influenza challenge compared to wild-type mice. This effect was not virus strain specific. Overall lung pathology and apoptosis were reduced in virus-infected KO mice. Production of proinflammatory cytokines and chemokines such as interleukin-10 (IL-10), gamma interferon (IFN-γ), and CC chemokine ligand 2 (CCL2) was also reduced in the lungs of the infected KO mice. Infiltration of neutrophils and depletion of CD11b+ macrophages, characteristic of severe influenza virus infection in mice, were lower in the KO animals. Together, these results demonstrate that activation of the P2X7 receptor is involved in the exacerbated immune response observed during influenza virus infection.IMPORTANCE A hallmark of influenza virus infection is the development of lung pathology induced by an exacerbated immune response. The mechanisms shared by the antiviral host defense required for viral clearance and those required for development of immunopathology are not clearly understood. Purinergic receptors, and in particular the purinergic receptor P2X7 (P2X7r), are involved in activation of the immune response. We used mice lacking the P2X7r (P2X7r KO mice) to better understand the mechanisms that lead to development of lung pathology during influenza virus infection. In our studies, we observed that P2X7r KO mice developed less lung immunopathology and had better survival than the wild-type mice. These results implicate P2X7r in the induction of an exacerbated local immune response to influenza virus and help us to better understand the mechanisms leading to the lung immunopathology observed during severe viral infections.


Asunto(s)
Interacciones Huésped-Patógeno , Pulmón/patología , Infecciones por Orthomyxoviridae/patología , Orthomyxoviridae/patogenicidad , Receptores Purinérgicos P2X7/metabolismo , Animales , Peso Corporal , Citocinas/análisis , Modelos Animales de Enfermedad , Macrófagos/inmunología , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/virología , Análisis de Supervivencia
15.
J Mol Biol ; 426(6): 1308-21, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24380762

RESUMEN

We previously demonstrated that ectodomain residue Asp286 in N2 neuraminidase (NA; Asp268 in N1 NA) present in budding-capable NA proteins contributes to productive NA plasma membrane transport partly by mediating escape from tetherin restriction [Yondola MA, Fernandes F, Belicha-Villanueva A, Uccelini M, Gao Q, Carter C, et al. (2011). Budding capability of the influenza virus neuraminidase can be modulated by tetherin. J Virol, 85, 2480-2491]. Budding-incapable NA proteins contain a G at this position and either co-expression of human immunodeficiency virus type 1 vpu or siRNA-mediated depletion of tetherin rescued budding capabilities in these proteins [Yondola MA, Fernandes F, Belicha-Villanueva A, Uccelini M, Gao Q, Carter C, et al. (2011). Budding capability of the influenza virus neuraminidase can be modulated by tetherin. J Virol, 85, 2480-2491]. Furthermore, replacement of D286 with G in budding-capable NA proteins caused loss of function, preventing release of NA virus-like particles (VLPs). Here, we show that mutation of this residue specifically modulates the ability of NA to escape tetherin restriction at the plasma membrane and results in virus attenuation in vivo. Based on immunogold electron microscopy and co-immunoprecipitation assays, both NAD286-containing and NAD286G-containing proteins associated with tetherin in the endoplasmic reticulum (ER). However, the NAD286G loss-of-function mutant also associated with the host factor outside the ER and in plasma-membrane-localized VLPs as visualized using immunogold electron microscopy. We conclude that the presence of aspartate at residue 286 liberates NA from tetherin-dependent restriction upon exit from the ER compartment thus preventing restriction at the plasma membrane. Underscoring the importance of these observations, knockdown of tetherin resulted in a 1-1.5 log increase in influenza virus growth. Additionally, the loss-of-function mutation conferred attenuation in a mouse model of influenza infection as evidenced by a 5-fold increase in LD50 and increases in either percent survival or time to death dependent on the administered dose in vivo.


Asunto(s)
Antígenos CD/metabolismo , Virus de la Influenza A/patogenicidad , Neuraminidasa/metabolismo , Infecciones por Orthomyxoviridae/virología , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Animales , Antígenos CD/genética , Células COS , Chlorocebus aethiops , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Estructura Terciaria de Proteína , Proteínas Virales/genética , Virión/fisiología
16.
Nat Commun ; 4: 2854, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24326875

RESUMEN

Without baseline human immunity to the emergent avian influenza A(H7N9) virus, neuraminidase inhibitors are vital for controlling viral replication in severe infections. An amino acid change in the viral neuraminidase associated with drug resistance, NA-R292K (N2 numbering), has been found in some H7N9 clinical isolates. Here we assess the impact of the NA-R292K substitution on antiviral sensitivity and viral replication, pathogenicity and transmissibility of H7N9 viruses. Our data indicate that an H7N9 isolate encoding the NA-R292K substitution is highly resistant to oseltamivir and peramivir and partially resistant to zanamivir. Furthermore, H7N9 reassortants with and without the resistance mutation demonstrate comparable viral replication in primary human respiratory cells, virulence in mice and transmissibility in guinea pigs. Thus, in stark contrast to oseltamivir-resistant seasonal influenza A(H3N2) viruses, H7N9 virus replication and pathogenicity in these models are not substantially altered by the acquisition of high-level oseltamivir resistance due to the NA-R292K mutation.


Asunto(s)
Farmacorresistencia Viral , Inhibidores Enzimáticos/farmacología , Subtipo H7N9 del Virus de la Influenza A/enzimología , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Neuraminidasa/metabolismo , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Animales , Antivirales/farmacología , Femenino , Humanos , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Humana/transmisión , Ratones , Ratones Endogámicos BALB C , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/genética , Oseltamivir/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Virulencia/efectos de los fármacos , Replicación Viral
17.
J Neuroimmunol ; 211(1-2): 73-83, 2009 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-19410300

RESUMEN

Influenza virus invades the olfactory bulb (OB) and enhances cytokine mRNAs therein at the time of illness onset. Here we show that viral antigen immunoreactivity co-localized with glial markers in the OB but could not be detected in other brain areas. Interleukin 1beta- and tumor necrosis factor alpha-immunoreactivity co-localized with neuronal markers in olfactory and central autonomic systems, and the number of cytokine-immunoreactive neurons increased at the time of illness onset [15 h post-inoculation (PI)] but not before (10 h PI). These results suggest that the OB virus influences the brain cytokines and therefore the onset of illness.


Asunto(s)
Sistema Nervioso Autónomo/inmunología , Citocinas/inmunología , Bulbo Olfatorio/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Sistema Nervioso Autónomo/metabolismo , Sistema Nervioso Autónomo/virología , Citocinas/metabolismo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Virus de la Influenza A , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Neuroglía/inmunología , Neuroglía/metabolismo , Neuroglía/virología , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/virología , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/virología , Infecciones por Orthomyxoviridae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA