Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Biomater Sci ; 8(8): 2283-2288, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32163067

RESUMEN

Highly efficient tumor-targeted therapy remains a great challenge due to the complexity and heterogeneity of tumor tissues. Herein, we developed an in vivo two-step tumor-targeting strategy by combining metabolic lipid-engineering with a stain-promoted azide-alkyne 1,3-dipolar cycloaddition (SPAAC) reaction, independent of the tumor microenvironment and cell phenotype. Firstly, exogenously-supplied azidoethyl-cholines (AECho) were metabolically incorporated into the cell membranes in tumor tissues through the intrinsic biosynthesis of phosphatidylcholine. The pre-inserted and accumulated azido groups (N3) could subsequently serve as 'artificial chemical receptors' for the specific anchoring of dibenzocyclooctyne (DBCO) modified biomimetic nanoparticles (DBCO-RBCG@ICG) via in situ click chemistry, resulting in significantly enhanced tumor-targeting and then an improved photothermal therapy effect. Such a two-step targeting strategy based on these cutting-edge techniques provided new insights into the universal and precise functionalization of living tissues for site-specific drug delivery in the diagnosis and treatment of various diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos , Membrana Eritrocítica , Animales , Línea Celular Tumoral , Colina/administración & dosificación , Colina/química , Química Clic , Colorantes/administración & dosificación , Colorantes/química , Humanos , Verde de Indocianina/administración & dosificación , Verde de Indocianina/química , Lípidos/administración & dosificación , Lípidos/química , Ingeniería Metabólica , Ratones , Neoplasias/terapia , Fototerapia
3.
Cell Death Dis ; 10(6): 442, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31165725

RESUMEN

We previously demonstrated that avian influenza A H7N9 virus preferentially infected CD14+ monocyte in human peripheral blood mononuclear cells (PBMCs), which led to apoptosis. To better understand H7N9 pathogenesis in relation to monocyte cell death, we showed here that extensive phosphorylation of mixed lineage kinase domain-like (MLKL) protein occurred concurrently with the activation of caspases-8, -9 and -3 in H7N9-infected monocytes at 6 h post infection (hpi), indicating that apoptosis and necroptosis pathways were simultaneously activated. The apoptotic morphology was readily observed in H7N9-infected monocytes with transmission electron microscopy (TEM), while the pan-caspase inhibitor, IDN6556 (IDN), accelerated cell death through necroptosis as evidenced by the increased level of pMLKL accompanied with cell swelling and plasma membrane rupture. Most importantly, H7N9-induced cell death could only be stopped by the combined treatment of IDN and necrosulfonamide (NSA), a pMLKL membrane translocation inhibitor, but not by individual inhibition of caspase or RIPK3. Our data further showed that activation of apoptosis and necroptosis pathways in monocytes differentially contributed to the immune response of monocytes upon H7N9 infection. Specifically, caspase inhibition significantly enhanced, while RIPK3 inhibition reduced the early expression of type I interferons and cytokine/chemokines in H7N9-infected monocytes. Moreover, culture supernatants from IDN-treated H7N9-infected monocyte promoted the expression of co-stimulatory molecule CD80, CD83 and CD86 on freshly isolated monocytes and monocyte-derived dendritic cells (MDCs) and enhanced the capacity of MDCs to induce CD3+ T-cell proliferation in vitro. In contrast, these immune stimulatory effects were abrogated by using culture supernatants from H7N9-infected monocyte with RIPK3 inhibition. In conclusion, our findings indicated that H7N9 infection activated both apoptosis and necroptosis in monocytes. An intact RIPK3 activity is required for upregulation of innate immune responses, while caspase activation suppresses the immune response.


Asunto(s)
Apoptosis/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Monocitos/inmunología , Monocitos/virología , Necroptosis/inmunología , Acrilamidas/farmacología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/genética , Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Inhibidores de Caspasas/farmacología , Citocinas/metabolismo , Células Dendríticas/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Interferón Tipo I/metabolismo , Monocitos/efectos de los fármacos , Monocitos/ultraestructura , Necroptosis/efectos de los fármacos , Fosforilación , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Sulfonamidas/farmacología , Linfocitos T/inmunología
4.
J Infect Dis ; 219(5): 795-807, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30202973

RESUMEN

BACKGROUND: Obesity is associated with increased severity of influenza infection. However, the underlying mechanism is largely unknown. METHODS: We employed a mouse model with diet-induced obesity (DIO) to study the innate immune responses induced by influenza virus. RESULTS: The lungs of DIO mice were heavily affected by obesity-associated chronic systemic inflammation with a significant increase in inflammatory cytokines/chemokines. Concurrently, lipid immune mediator prostaglandin E2 (PGE2) was also significantly elevated in DIO mice. However, the DIO mice mounted a blunted and delayed upregulation of mRNA and protein concentrations of interferon-ß and inflammatory cytokines/chemokines upon A(H1N1)pdm09 virus (H1N1/415742Md) challenge compared with those of lean mice. PGE2 concentrations were significantly higher in the lungs of DIO mice compared to that of lean mice postchallenge. Treatment with paracetamol in challenged DIO mice significantly enhanced the expression of interferon-α/ß and cytokine genes at days 1 and 3 postinfection compared with that of untreated DIO mice. Furthermore, paracetamol treatment alone started 3 days before virus challenge and continued until 6 days postchallenge ameliorated the severity of a lethal H1N1/415742Md infection in DIO mice with improved survival. CONCLUSIONS: Impaired innate response to influenza in DIO mice is associated with elevated PGE2, which could be restored to some degree by paracetamol treatment.


Asunto(s)
Acetaminofén/administración & dosificación , Dinoprostona/metabolismo , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/administración & dosificación , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Citocinas/metabolismo , Femenino , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Obesos , Infecciones por Orthomyxoviridae/patología , Resultado del Tratamiento
5.
Front Immunol ; 9: 2370, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30369932

RESUMEN

Current influenza vaccines have relatively low effectiveness, especially against antigenically drifted strains, the effectiveness is even lower in the elderly and immunosuppressed individuals. We have previously shown in a randomized clinical trial that the topical application of a toll-like receptor 7 agonist, imiquimod, just before intradermal influenza vaccine could expedite and augment antibody response, including to antigenically-drifted strains. However, the mechanism of this vaccine and imiquimod combination approach is poorly understood. Here, we demonstrated that imiquimod alone directly activated purified mouse peritoneal B cells. When combined with inactivated H1N1/415742Md influenza virus particle (VP) as vaccine, co-stimulation of mouse peritoneal B cells in vitro induced stronger activation, proliferation, and production of virus-antigen specific IgM and IgG. Intraperitoneal injection of a combination of VP and imiquimod (VCI) was associated with an increased number of activated B cells with enhanced expression of CD86 in the mesenteric draining lymph nodes (mesLN) and the spleen at 18 h after injection. Three days after immunization with VCI, mouse spleen showed significantly more IgM and IgG secreting cells upon in vitro re-stimulation with inactivated virus, mouse sera were detected with viral neutralizing antibody. Transfer of these spleen B cells to naïve mice improved survival after lethal dose of H1N1/415742Md challenge. More importantly, the functional response of VCI-induced B cell activation was demonstrated by early challenge with a lethal dose of H1N1/415742Md influenza virus at 3 days after immunization. The spleen and mediastinal lymph nodes (mdLN) in mice immunized with VCI had germinal center formation, and significantly higher number of plasmablasts, plasma cells, and virus-antigen specific IgM and IgG secreting cells at only 3-4 days post virus challenge, compared with those of mice that have received imiquimod, inactivated virus alone or PBS. Serum virus-specific IgG2a, IgG2b, and IgG1 and bronchoalveolar lavage fluid (BALF) virus-specific IgA at 3 or 4 days post challenge were significantly higher in mice immunized with VCI, which had significantly reduced lung viral load and 100% survival. These findings suggested that imiquimod accelerates the vaccine-induced antibody production via inducing rapid differentiation of naïve B cells into antigen-specific antibody producing cells.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Virus de la Influenza A/inmunología , Activación de Linfocitos/inmunología , Receptor Toll-Like 7/metabolismo , Animales , Anticuerpos Antivirales/inmunología , Antígenos/inmunología , Apoptosis , Diferenciación Celular/inmunología , Femenino , Humanos , Inmunización , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Bazo/inmunología , Bazo/metabolismo
6.
Arch Virol ; 163(9): 2349-2358, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29736671

RESUMEN

Seasonal influenza virus remains a common cause of mortality despite the use of neuraminidase inhibitors. This study evaluated the efficacy of a triple combination of zanamivir, clarithromycin and flufenamic acid (FFA) in the treatment of influenza virus A(H1N1) infection. An in vitro cell protection assay and a multiple-cycle growth assay showed that the antiviral activity of zanamivir was enhanced when combined with clarithromycin or FFA. A mouse challenge model was used here for the evaluation of the in vivo efficacy of the triple combination treatment. We found that mice receiving the triple combination of FFA, zanamivir, and clarithromycin had a significantly better survival rate than those receiving the double combination of zanamivir and clarithromycin (88% versus 44%, P = 0.0083) or zanamivir monotherapy (88% versus 26%, P = 0.0002). Mice in the FFA-zanamivir-clarithromycin triple combination group also exhibited significantly less body weight loss than those in the zanamivir-clarithromycin double combination group. There was no significant difference in the lung viral titers among the different groups from day 2 to day 6 postinfection. However, the levels of IL-1ß, TNF-α and RANTES in the FFA-zanamivir-clarithromycin triple combination group were significantly lower than those in the zanamivir-clarithromycin double combination group, zanamivir monotherapy group, or solvent group on day 2 postinfection. Our findings showed that the FFA-zanamivir-clarithromycin triple combination improved the inflammatory markers and survival of severe influenza A(H1N1) infection in mice.


Asunto(s)
Antivirales/administración & dosificación , Claritromicina/administración & dosificación , Ácido Flufenámico/administración & dosificación , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Gripe Humana/mortalidad , Zanamivir/administración & dosificación , Animales , Aprobación de Drogas/legislación & jurisprudencia , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/virología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Estados Unidos , United States Food and Drug Administration
7.
Emerg Microbes Infect ; 7(1): 23, 2018 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-29511175

RESUMEN

The 2017 Hong Kong influenza A(H3N2) summer season was unexpectedly severe. However, antigenic characterization of the 2017 circulating A(H3N2) viruses using ferret antisera did not show significant antigenic drift. We analyzed the hemagglutinin amino acid sequences of A(H3N2) virus circulating in Hong Kong in 2017, and found that viruses with hemagglutinin N121K substitution, which was rare before 2017, emerged rapidly and dominated in 2017 (52.4% of A[H3N2] virus in 2017 contains N121K substitution). Microneutralization assay using archived human sera collected from mid-2017 showed that the geometric mean microneutralization titer was 3.6-fold lower against a 2017 cell culture-grown circulating A(H3N2)-N121K virus (3391/2017 virus) than that against the cell culture-grown 2016-2017 A(H3N2) seasonal influenza vaccine-like vaccine virus (4801/2014 virus) (13.4 vs 41.8, P < 0.0001). Significantly fewer serum specimens had a microneutralization titer of 40 or above against 3391/2017 virus than that against 4801/2014 virus (26.4% vs 60.0%, P < 0.0001). Conversely, the geometric mean hemagglutination inhibition titer was slightly higher against 3391/2017 virus than that against the 4801/2014 virus (96.9 vs 55.4, P < 0.0001). Moreover, 59.1% of specimens had a significantly lower microneutralization antibody titer (≥4-fold) against 3391/2017 virus than that against 4801/2014 virus, but none for hemagglutination titer (P < 0.0001). Similar results of microneutralization and hemagglutination titers were observed for day 21-post-vaccination sera. Hence, the 2017 A(H3N2) summer peak in Hong Kong was associated with a low-microneutralization titer against the circulating virus. Our results support the use of microneutralization assay with human serum in assessing population susceptibility and antigenic changes of A(H3N2) virus. Novel and available immunization approach, such as topical imiquimod followed by intradermal vaccination, to broaden the neutralizing antibody response of influenza vaccine should be considered.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Subtipo H3N2 del Virus de la Influenza A/inmunología , Gripe Humana/sangre , Adolescente , Adulto , Anciano , Femenino , Hong Kong/epidemiología , Humanos , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Gripe Humana/epidemiología , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Filogenia , Estaciones del Año , Vacunación , Adulto Joven
8.
Anal Chem ; 89(21): 11620-11627, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-28971673

RESUMEN

Though techniques in bioorthogonal chemistry and metabolic incorporation have been developed over the past decade, it remains difficult to integrate different bioorthogonal reactions or metabolic incorporations into one system. In this report, the protein and DNA metabolic incorporations were combined with two bioorthogonal reactions in one cell to develop a facile and universal method for virus dual labeling. Azide and vinyl groups were introduced into the proteins or genomes of viruses, respectively, through the intrinsic biosynthesis of biomolecules, which were subsequently fluorescently labeled via copper-free click chemistry or alkene-tetrazine ligation reactions during natural propagation process in host cells. Both the envelope viruses and the capsid viruses could be labeled, and the dual labeling efficiency was more than 80%. The labeled progeny virions were structurally intact and fully infectious, and their fluorescence was strong enough to track single virions.


Asunto(s)
Coloración y Etiquetado/métodos , Virus Vaccinia/metabolismo , Alanina/análogos & derivados , Alanina/metabolismo , Animales , Cápside/metabolismo , Chlorocebus aethiops , Química Clic , ADN Viral/química , ADN Viral/metabolismo , Colorantes Fluorescentes/metabolismo , Conformación de Ácido Nucleico , Células Vero
9.
J Gen Virol ; 98(5): 922-934, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28555541

RESUMEN

Most patients with avian influenza A H7N9 virus (H7N9) infection suffer from severe illness, accompanied by dysregulated cytokine/chemokine response, delayed viral clearance and impaired neutralizing antibody response. Here, we evaluated the role of peripheral blood mononuclear cells (PBMCs) in the pathogenesis of H7N9 infection using an ex vivo infection model. H7N9 infected a significantly higher percentage of PBMCs (23.9 %) than those of avian influenza A H5N1 virus (H5N1) (12.3 %) and pandemic H1N1 virus (pH1N1) (5.5 %) (P<0.01). H7N9 infected significantly more B and T lymphocytes than H5N1. When compared with pH1N1, H7N9-infected PBMCs had significantly higher mRNA levels of proinflammatory cytokines and type I interferons (IFNs) at 6 h post-infection (p.i.), but significantly lower levels of IFN-γ and IP-10 at 12 h p.i. Among the PBMCs, CD14+ monocytes were most permissive to H7N9 infection. The percentage of infected CD14+ monocytes was significantly higher for H7N9 than that of pH1N1, but not significantly different from that of H5N1. H7N9-infected monocytes showed higher expression of MIP-1α, MIP-1ß and RANTES than that of pH1N1 at 6 h p.i. H7N9- but not pH1N1-infected monocytes died rapidly via apoptosis. Furthermore, pH1N1- but not H7N9-infected monocytes showed increased expression of the monocyte activation and differentiation markers. Unlike pH1N1, H7N9 showed similar PBMC/monocyte cytokine/chemokine expression profile, monocyte cell death and expression of activation/differentiation markers to H5N1. Besides proinflammatory cytokine activation leading to a cytokine storm, impaired IFN-γ production, rapid monocytic death and lack of monocyte differentiation may affect the ability of H7N9-infected innate immune cells to recruit protective adaptive immunity.


Asunto(s)
Apoptosis , Citocinas/metabolismo , Subtipo H7N9 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H7N9 del Virus de la Influenza A/inmunología , Leucocitos Mononucleares/virología , Células Cultivadas , Humanos , Subtipo H7N9 del Virus de la Influenza A/patogenicidad
10.
EBioMedicine ; 14: 112-122, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27884655

RESUMEN

BACKGROUND: Disseminated or fatal Zika virus (ZIKV) infections were reported in immunosuppressed patients. Existing interferon-signaling/receptor-deficient mouse models may not be suitable for evaluating treatment effects of recombinant interferons. METHODS: We developed a novel mouse model for ZIKV infection by immunosuppressing BALB/c mice with dexamethasone. RESULTS: Dexamethasone-immunosuppressed male mice (6-8weeks) developed disseminated infection as evidenced by the detection of ZIKV-NS1 protein expression and high viral loads in multiple organs. They had ≥10% weight loss and high clinical scores soon after dexamethasone withdrawal (10dpi), which warranted euthanasia at 12dpi. Viral loads in blood and most tissues at 5dpi were significantly higher than those at 12dpi (P<0.05). Histological examination revealed prominent inflammatory infiltrates in multiple organs, and CD45+ and CD8+ inflammatory cells were seen in the testis. These findings suggested that clinical deterioration occurred during viral clearance by host immune response. Type I interferon treatments improved clinical outcome of mice (100% vs 0% survival). CONCLUSIONS: Besides virus dissemination, inflammation of various tissues, especially orchitis, may be potential complications of ZIKV infection with significant implications on disease transmission and male fertility. Interferon treatment should be considered in patients at high risks for ZIKV-associated complications when the potential benefits outweigh the side effects of treatment.


Asunto(s)
Antivirales/farmacología , Huésped Inmunocomprometido , Interferón Tipo I/farmacología , Orquitis/virología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Virus Zika/efectos de los fármacos , Virus Zika/inmunología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Dexametasona/efectos adversos , Modelos Animales de Enfermedad , Femenino , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Orquitis/tratamiento farmacológico , Carga Viral , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/patología
11.
Clin Vaccine Immunol ; 22(12): 1235-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26446420

RESUMEN

Influenza A(H7N9) virus pneumonia is associated with a high case fatality rate in humans. Multiple viral factors have been postulated to account for the high virulence of the virus. It has been reported that patients with influenza A(H7N9) virus infection have relatively low titers of neutralizing antibodies compared to those with seasonal influenza virus infections. In this study, we compared serum hemagglutination inhibition (HI) and microneutralization (MN) antibody titers of mice challenged with wild-type A(H7N9) viruses [H7N9(Anhui) and H7N9(Zhejiang)], an A(H1N1)pdm09 virus [pH1N1(2009)], and a recombinant A(H7N9) virus with PR8/H1N1 internal genes (rg-PR8-H7-N9). All mice infected by H7N9(Anhui) and H7N9(Zhejiang) developed serum HI antibodies at 14 days postinfection (dpi) but no detectable MN antibodies, even at 28 dpi. A low level of neutralizing activity was detected in H7N9(Anhui)- and H7N9(Zhejiang)-infected mice using fluorescent focus MN assay, but convalescent-phase serum samples obtained from H7N9(Anhui)-infected mice did not reduce the mortality of naive mice after homologous virus challenge. Reinfection with homologous A(H7N9) virus induced higher HI and MN titers than first infection. In contrast, pH1N1(2009) virus infection induced robust HI and MN antibody responses, even during the first infection. Moreover, rg-PR8-H7-N9 induced significantly higher HI and MN antibody titers than H7N9(Zhejiang). In conclusion, the internal genes of A(H7N9) virus can affect the humoral immune response against homologous viral surface proteins, which may also contribute to the virulence of A(H7N9) virus.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Genes Virales , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Protección Cruzada , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunidad Humoral , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Proteínas de la Matriz Viral/inmunología
12.
PLoS One ; 9(9): e107966, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25232731

RESUMEN

BACKGROUND: Human infection caused by the avian influenza A H7N9 virus has a case-fatality rate of over 30%. Systematic study of the pathogenesis of avian H7N9 isolate and effective therapeutic strategies are needed. METHODS: BALB/c mice were inoculated intranasally with an H7N9 virus isolated from a chicken in a wet market epidemiologically linked to a fatal human case, (A/chicken/Zhejiang/DTID-ZJU01/2013 [CK1]), and with an H7N9 virus isolated from a human (A/Anhui/01/2013 [AH1]). The pulmonary viral loads, cytokine/chemokine profiles and histopathological changes of the infected mice were compared. The therapeutic efficacy of a non-steroidal anti-inflammatory drug (NSAID), celecoxib, was assessed. RESULTS: Without prior adaptation, intranasal inoculation of 106 plaque forming units (PFUs) of CK1 caused a mortality rate of 82% (14/17) in mice. Viral nucleoprotein and RNA expression were limited to the respiratory system and no viral RNA could be detected from brain, liver and kidney tissues. CK1 caused heavy alveolar inflammatory exudation and pulmonary hemorrhage, associated with high pulmonary levels of proinflammatory cytokines. In the mouse lung cell line LA-4, CK1 also induced high levels of interleukin-6 (IL-6) and cyclooxygenase-2 (COX-2) mRNA. Administration of the antiviral zanamivir did not significantly improve survival in mice infected with CK1, but co-administration of the non-steroidal anti-inflammatory drug (NSAID) celecoxib in combination with zanamivir improved survival and lung pathology. CONCLUSIONS: Our findings suggested that H7N9 viruses isolated from chicken without preceding trans-species adaptation can cause lethal mammalian pulmonary infection. The severe proinflammatory responses might be a factor contributing to the mortality. Treatment with combination of antiviral and NSAID could ameliorate pulmonary inflammation and may improve survival.


Asunto(s)
Antivirales/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Subtipo H7N9 del Virus de la Influenza A/fisiología , Neumonía Viral/tratamiento farmacológico , Zanamivir/farmacología , Adaptación Fisiológica/inmunología , Animales , Antivirales/uso terapéutico , Línea Celular , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Humanos , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Subtipo H7N9 del Virus de la Influenza A/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Neumonía Viral/inmunología , Neumonía Viral/virología , Replicación Viral , Zanamivir/uso terapéutico
13.
Clin Infect Dis ; 59(9): 1246-55, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25048848

RESUMEN

BACKGROUND: Imiquimod, a synthetic Toll-like receptor 7 agonist enhanced immunogenicity of influenza vaccine in a mouse model. We hypothesized that topical imiquimod before intradermal influenza vaccination (TIV) would produce similar effect in human. METHODS: We performed a prospective 1-year follow-up, double-blind, randomized, controlled trial with adults with comorbidities. Participants were randomized to 1 of the following 3 vaccinations: topical 5% 250 mg imiquimod ointment followed by intradermal TIV, topical aqueous-cream followed by intradermal TIV, or topical aqueous-cream followed by intramuscular TIV. Patients and investigators were blinded to the type of topical treatment applied. Hemagglutination inhibition (HI) and microneutralization antibody titers were measured. The primary outcome was the day 7 seroconversion rate. RESULTS: Ninety-one recruited participants completed the study. The median age was 73 years. On day 7, 27/30 (90%) patients who received imiquimod and intradermal TIV achieved seroconversion against the H1N1 strain by HI, compared with 4/30 (13.3%) who received aqueous-cream and intramuscular TIV (P < .001), and 12/31 (38.7%) who received aqueous-cream and intradermal TIV (P < .001). The seroconversion, seroprotection, and geometric mean titer-fold increase were met in all 3 strains in the imiquimod and intradermal TIV group 2 weeks earlier, and the better seroconversion rate was sustained from day 7 to year 1 (P ≤ .001). The better immunogenicity was associated with fewer hospitalizations for influenza or pneumonia (P < .05). All adverse reactions were self-limited. CONCLUSIONS: Pretreatment with topical imiquimod significantly expedited, augmented, and prolonged the immunogenicity of influenza vaccination. This strategy for influenza immunization should be considered for the elderly population.


Asunto(s)
Aminoquinolinas/administración & dosificación , Aminoquinolinas/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Administración Tópica , Anciano , Anciano de 80 o más Años , Aminoquinolinas/efectos adversos , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Imiquimod , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/química , Inyecciones Intradérmicas , Masculino , Persona de Mediana Edad
14.
Clin Vaccine Immunol ; 21(4): 570-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24521786

RESUMEN

Toll-like receptors (TLRs) of the innate immune system are known targets for enhancing vaccine efficacy. We investigated whether imiquimod, a synthetic TLR7 agonist, can expedite the immune response against influenza virus infection when combined with influenza vaccine. BALB/c mice were immunized intraperitoneally with monovalent A(H1N1)pdm09 vaccine combined with imiquimod (VCI) prior to intranasal inoculation with a lethal dose of mouse-adapted A(H1N1)pdm09 virus. For mice immunized 3 days before infection, the survival rates were significantly higher in the VCI group (60%, mean survival time[MST], 11 days) than in the vaccine-alone (30%; MST, 8.8 days), imiquimod-alone (5%; MST, 8.4 days), and phosphate-buffered saline (PBS) (0%; MST, 6.2 days) groups (P < 0.01). In the VCI group, 45 and 35% of the mice survived even when they were infected 2 days or 1 day after immunization. Virus-specific serum IgM, IgG, and neutralizing antibodies appeared earlier with higher geometric mean titers in the VCI group than in the control groups. The pulmonary viral load was significantly lower at all time points postinfection in the VCI, vaccine-alone, and imiquimod-alone groups than in the PBS control group (P < 0.05). The protection induced by VCI was specific for A(H1N1)pdm09 virus but not for A(H5N1) virus. Since imiquimod combined with RNase-treated vaccine is as protective as imiquimod combined with untreated vaccine, mechanisms other than TLR7 may operate in expediting and augmenting immune protection. Moreover, increased gamma interferon mRNA expression and IgG isotype switching, which are markers of the Th1 response induced by imiquimod, were not apparent in our mouse model. The mechanisms of imiquimod-induced immune protection deserve further study.


Asunto(s)
Aminoquinolinas/administración & dosificación , Anticuerpos Antivirales/sangre , Factores Inmunológicos/administración & dosificación , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Femenino , Imiquimod , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Glicoproteínas de Membrana/agonistas , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Análisis de Supervivencia , Receptor Toll-Like 7/agonistas
15.
Biotechnol Lett ; 34(3): 433-40, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22105552

RESUMEN

A new line of outer root sheath (ORS) cells was established from hair follicles of Jining grey goat by using a mechanical separation combined with enzyme digestion. Cell morphology is described at different phases. The chromosome analysis of ORS cells, identification of the ORS cells and morphological reversion test were detected at the 4th and 40th passages. The ORS cells were healthy and the growth characteristics were stable with a population doubling time of 52 h. Chromosome analysis showed that >58% of cells were diploid. Test for ORS cell line CK19 expression was positive. This newly established ORS cell line not only lays the foundation for further studying on the growth, regeneration, development law of goat hair follicle but also provides a mirror for the research of human hair in medical field.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Línea Celular , Cabras , Folículo Piloso/citología , Animales , Diploidia , Cariotipificación , Pase Seriado
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...