Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
N Engl J Med ; 383(5): 452-459, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32726531

RESUMEN

BACKGROUND: Insufficient vaccine doses and the lack of therapeutic agents for yellow fever put global health at risk, should this virus emerge from sub-Saharan Africa and South America. METHODS: In phase 1a of this clinical trial, we assessed the safety, side-effect profile, and pharmacokinetics of TY014, a fully human IgG1 anti-yellow fever virus monoclonal antibody. In a double-blind, phase 1b clinical trial, we assessed the efficacy of TY014, as compared with placebo, in abrogating viremia related to the administration of live yellow fever vaccine (YF17D-204; Stamaril). The primary safety outcomes were adverse events reported 1 hour after the infusion and throughout the trial. The primary efficacy outcome was the dose of TY014 at which 100% of the participants tested negative for viremia within 48 hours after infusion. RESULTS: A total of 27 healthy participants were enrolled in phase 1a, and 10 participants in phase 1b. During phase 1a, TY014 dose escalation to a maximum of 20 mg per kilogram of body weight occurred in 22 participants. During phases 1a and 1b, adverse events within 1 hour after infusion occurred in 1 of 27 participants who received TY014 and in none of the 10 participants who received placebo. At least one adverse event occurred during the trial in 22 participants who received TY014 and in 8 who received placebo. The mean half-life of TY014 was approximately 12.8 days. At 48 hours after the infusion, none of the 5 participants who received the starting dose of TY014 of 2 mg per kilogram had detectable YF17D-204 viremia; these participants remained aviremic throughout the trial. Viremia was observed at 48 hours after the infusion in 2 of 5 participants who received placebo and at 72 hours in 2 more placebo recipients. Symptoms associated with yellow fever vaccine were less frequent in the TY014 group than in the placebo group. CONCLUSIONS: This phase 1 trial of TY014 did not identify worrisome safety signals and suggested potential clinical benefit, which requires further assessment in a phase 2 trial. (Funded by Tysana; ClinicalTrials.gov number, NCT03776786.).


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Vacuna contra la Fiebre Amarilla , Fiebre Amarilla/tratamiento farmacológico , Virus de la Fiebre Amarilla/inmunología , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Semivida , Humanos , Estimación de Kaplan-Meier , Viremia/tratamiento farmacológico , Fiebre Amarilla/virología , Virus de la Fiebre Amarilla/efectos de los fármacos
2.
PLoS One ; 13(1): e0190063, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29293541

RESUMEN

Influenza and asthma are two of the major public health concerns in the world today. During the 2009 influenza pandemic asthma was found to be the commonest comorbid illness of patients admitted to hospital. Unexpectedly, it was also observed that asthmatic patients admitted to hospital with influenza infection were less likely to die or require admission to intensive care compared with non-asthmatics. Using an in vivo model of asthma and influenza infection we demonstrate that prior exposure to Blomia tropicalis extract (BTE) leads to an altered immune response to influenza infection, comprised of less severe weight loss and faster recovery following infection. This protection was associated with significant increases in T cell numbers in the lungs of BTE sensitised and infected mice, as well as increased IFN-γ production from these cells. In addition, elevated numbers of CD11b+ dendritic cells (DCs) were found in the lung draining lymph nodes following infection of BTE sensitised mice compared to infected PBS treated mice. These CD11b+ DCs appeared to be better at priming CD8 specific T cells both in vivo and ex vivo, a function not normally attributed to CD11b+ DCs. We propose that this alteration in cross-presentation and more efficient T cell priming seen in BTE sensitised mice, led to the earlier increase in T cells in the lungs and subsequently faster clearance of the virus and reduced influenza induced pathology. We believe this data provides a novel mechanism that explains why asthmatic patients may present with less severe disease when infected with influenza.


Asunto(s)
Alérgenos/administración & dosificación , Asma/inmunología , Células Dendríticas/inmunología , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T/inmunología , Animales , Asma/complicaciones , Líquido del Lavado Bronquioalveolar , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Exposición por Inhalación , Interferón gamma/biosíntesis , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/virología , Extractos Vegetales/farmacología , Carga Viral
3.
Front Immunol ; 8: 1484, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29163545

RESUMEN

CD8+ T cells play an important role in providing protective immunity against a wide range of pathogens, and a number of different factors control their activation. Although CD40L-mediated CD40 licensing of dendritic cells (DCs) by CD4+ T cells is known to be necessary for the generation of a robust CD8+ T cell response, the contribution of CD8+ T cell-expressed CD40L on DC licensing is less clear. We have previously shown that CD8+ T cells are able to induce the production of IL-12 p70 by DCs in a CD40L-dependent manner, providing some evidence that CD8+ T cell-mediated activation of DCs is possible. To better understand the role of CD40L on CD8+ T cell responses, we generated and characterized CD40L-expressing CD8+ T cells both in vitro and in vivo. We found that CD40L was expressed on 30-50% of effector CD8+ T cells when stimulated and that this expression was transient. The expression of CD40L on CD8+ T cells promoted the proliferation and differentiation of both the CD40L-expressing CD8+ T cells and the bystander effector CD8+ T cells. This process occurred via a cell-extrinsic manner and was mediated by DCs. These data demonstrate the existence of a mechanism where CD8+ T cells and DCs cooperate to maximize CD8+ T cell responses.

4.
Immunology ; 151(2): 227-238, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28190273

RESUMEN

Sensitization of allergic patients normally takes place over several years and is the result of repeated exposure to low levels of allergen. Most mouse asthma models use a high dose of allergen administered over a short period. We have investigated the role of dose in the immune response to an inhaled respiratory allergen (Blomia tropicalis). We observed the effect of priming dose on the allergic response in mice intranasally immunized with low (0·5 µg) and high (50 µg) doses of B. tropicalis extract and killed 1 day after the last challenge. For both doses of allergen, T helper type 2 (Th2) cells and Th2 cytokines were evident as well as eosinophilic inflammation accompanied by mucus hyper-secretion. By contrast, IgE and IgG1 antibody responses were normally only detected at high-dose priming. To investigate the mechanism for these effects, we found group 2 innate lymphoid cells (ILC2s) were increased 48 hr after challenge in the low-dose-treated but not the high-dose-treated mice. Furthermore, we determined whether repeated low-dose exposure with different priming protocols could induce an antibody response. Repeated low-dose exposure to 0·5 µg three times weekly for 4 weeks (cumulative 6 µg) had the same effect as a shorter high-dose exposure (cumulative 80 µg) and increasing cumulative dose induced antibody responses. These data indicate that low doses of allergen are sufficient to prime Th2 cells and ILC2s, but insufficient to induce antibody responses. Cumulative exposure to small amounts of allergen induces both Th2 and antibody responses and may better reflect natural sensitization.


Asunto(s)
Alérgenos/inmunología , Diferenciación Celular , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Activación de Linfocitos , Células Th2/citología , Células Th2/inmunología , Alérgenos/administración & dosificación , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Thorax ; 70(6): 528-36, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25841236

RESUMEN

BACKGROUND: Current data concerning maternal paracetamol intake during pregnancy, or intake during infancy and risk of wheezing or asthma in childhood is inconclusive based on epidemiological studies. We have investigated whether there is a causal link between maternal paracetamol intake during pregnancy and lactation and the development of house dust mite (HDM) induced allergic airways disease (AAD) in offspring using a neonatal mouse model. METHODS: Pregnant mice were administered paracetamol or saline by oral gavage from the day of mating throughout pregnancy and/or lactation. Subsequently, their pups were exposed to intranasal HDM or saline from day 3 of life for up to 6 weeks. Assessments of airway hyper-responsiveness, inflammation and remodelling were made at weaning (3 weeks) and 6 weeks of age. RESULTS: Maternal paracetamol exposure either during pregnancy and/or lactation did not affect development of AAD in offspring at weaning or at 6 weeks. There were no effects of maternal paracetamol at any time point on airway remodelling or IgE levels. CONCLUSIONS: Maternal paracetamol did not enhance HDM induced AAD in offspring. Our mechanistic data do not support the hypothesis that prenatal paracetamol exposure increases the risk of childhood asthma.


Asunto(s)
Acetaminofén/farmacología , Analgésicos no Narcóticos/farmacología , Hiperreactividad Bronquial , Exposición Materna , Animales , Animales Recién Nacidos , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Modelos Animales de Enfermedad , Femenino , Ratones , Embarazo
6.
J Virol ; 87(23): 12510-22, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24027334

RESUMEN

The factors that regulate the contraction of the CD8 T cell response and the magnitude of the memory cell population against localized mucosal infections such as influenza are important for generation of efficient vaccines but are currently undefined. In this study, we used a mouse model of influenza to demonstrate that the absence of gamma interferon (IFN-γ) or IFN-γ receptor 1 (IFN-γR1) leads to aberrant contraction of antigen-specific CD8 T cell responses. The increased accumulation of the effector CD8 T cell population was independent of viral load. Reduced contraction was associated with an increased fraction of CD8 T cells expressing the interleukin-7 receptor (IL-7R) at the peak of the response, resulting in enhanced numbers of memory/memory precursor cells in IFN-γ(-/-) and IFN-γR(-/-) compared to wild-type (WT) mice. Blockade of IL-7 within the lungs of IFN-γ(-/-) mice restored the contraction of influenza virus-specific CD8 T cells, indicating that IL-7R is important for survival and is not simply a consequence of the lack of IFN-γ signaling. Finally, enhanced CD8 T cell recall responses and accelerated viral clearance were observed in the IFN-γ(-/-) and IFN-γR(-/-) mice after rechallenge with a heterologous strain of influenza virus, confirming that higher frequencies of memory precursors are formed in the absence of IFN-γ signaling. In summary, we have identified IFN-γ as an important regulator of localized viral immunity that promotes the contraction of antigen-specific CD8 T cells and inhibits memory precursor formation, thereby limiting the size of the memory cell population after an influenza virus infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Interferón gamma/inmunología , Animales , Linfocitos T CD8-positivos/citología , Femenino , Humanos , Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/virología , Interferón gamma/deficiencia , Interferón gamma/genética , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/inmunología , Especificidad de la Especie , Receptor de Interferón gamma
7.
J Virol ; 84(17): 8790-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20573822

RESUMEN

Regulatory CD4(+) T cells have been shown to be important in limiting immune responses, but their role in respiratory viral infections has received little attention. Here we observed that following respiratory syncytial virus (RSV) infection, CD4(+) Foxp3(+) CD25(+) natural regulatory T-cell numbers increased in the bronchoalveolar lavage fluid, lung, mediastinal lymph nodes, and spleen. The depletion of CD25(+) natural regulatory T cells prior to RSV infection led to enhanced weight loss with delayed recovery that was surprisingly accompanied by increased numbers of activated natural killer cells in the lung and bronchoalveolar lavage fluid on day 8 postinfection. Increased numbers of neutrophils were also detected within the bronchoalveolar lavage fluid and correlated with elevated levels of myeloperoxidase as well as interleukin-6 (IL-6) and gamma interferon (IFN-gamma). CD25(+) natural regulatory T-cell depletion also led to enhanced numbers of proinflammatory T cells producing IFN-gamma and tumor necrosis factor alpha (TNF-alpha) in the lung. Despite these increases in inflammatory responses and disease severity, the viral load was unaltered. This work highlights a critical role for natural regulatory T cells in regulating the adaptive and innate immune responses during the later stages of lung viral infections.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/fisiología , Linfocitos T Reguladores/inmunología , Animales , Línea Celular , Femenino , Humanos , Interferón gamma/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Interleucina-6/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/inmunología , Linfocitos T Reguladores/virología
8.
J Virol ; 84(10): 5294-302, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20200251

RESUMEN

Respiratory syncytial virus (RSV) is the main cause of bronchiolitis, the major cause of hospitalization of infants. An ideal RSV vaccine would be effective for neonates, but the immune responses of infants differ markedly from those of adults, often showing a bias toward T-helper 2 (Th2) responses and reduced gamma interferon (IFN-gamma) production. We previously developed recombinant RSV vectors expressing IFN-gamma and interleukin-4 (IL-4) that allow us to explore the role of these key Th1 and Th2 cytokines during infection. The aim of the current study was to explore whether an immunomodulation of infant responses could enhance protection. The expression of IFN-gamma by a recombinant RSV vector (RSV/IFN-gamma) attenuated primary viral replication in newborn mice without affecting the development of specific antibody or T-cell responses. Upon challenge, RSV/IFN-gamma mice were protected from the exacerbated disease observed for mice primed with wild-type RSV; however, antiviral immunity was not enhanced. Conversely, the expression of IL-4 by recombinant RSV did not affect virus replication in neonates but greatly enhanced Th2 immune responses upon challenge without affecting weight loss. These studies demonstrate that it is possible to manipulate infant immune responses by using cytokine-expressing recombinant viruses and that neonatal deficiency in IFN-gamma responses may lead to enhanced disease during secondary infection.


Asunto(s)
Interferón gamma/inmunología , Pulmón/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Anticuerpos Antivirales/sangre , Peso Corporal , Femenino , Interferón gamma/genética , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Embarazo , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Vacunas contra Virus Sincitial Respiratorio/genética , Virus Sincitiales Respiratorios/genética , Linfocitos T/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
9.
J Virol ; 84(8): 4073-82, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20130064

RESUMEN

Respiratory syncytial virus (RSV) causes bronchiolitis, the main cause of infantile hospitalization. Immunity against reinfection is poor, and there is great interest in boosting vaccine responses using live vectors expressing host cytokines. We therefore constructed a recombinant RSV expressing murine interleukin 18 (RSV/IL-18), a cytokine capable of inducing strong antiviral immune responses. In vitro RSV/IL-18 replicated at wild-type levels and produced soluble IL-18. In naïve BALB/c mice, RSV/IL-18 infection significantly increased both IL-18 mRNA and protein and attenuated the peak viral load 3-fold. Despite a reduced viral load, RSV/IL-18 infection caused a biphasic weight loss at days 2 and 6 postinfection that was not seen in wild-type infection. Day 2 disease was associated with enhanced pulmonary natural killer (NK) cell numbers and activity and was prevented by NK cell depletion during infection; day 6 disease was correlated with CD8 T-cell recruitment and was enhanced by NK cell depletion. IL-18 expression during priming also enhanced RSV-specific antibody responses and T-cell responses on secondary RSV infection. Therefore, while IL-18 boosted antiviral immunity and reduced the viral load, its coexpression worsened disease. This is the first recombinant RSV with this property, and these are the first studies to demonstrate that NK cells can induce pathology during pulmonary viral infections.


Asunto(s)
Interleucina-18/inmunología , Células Asesinas Naturales/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/inmunología , Animales , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Femenino , Interleucina-18/genética , Procedimientos de Reducción del Leucocitos , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/genética , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/genética , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Viremia , Pérdida de Peso
10.
Nat Med ; 11(4): 429-33, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15768029

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease resulting in the selective death of motor neurons in the brain and spinal cord. Some familial cases of ALS are caused by dominant mutations in the gene encoding superoxide dismutase (SOD1). The emergence of interfering RNA (RNAi) for specific gene silencing could be therapeutically beneficial for the treatment of such dominantly inherited diseases. We generated a lentiviral vector to mediate expression of RNAi molecules specifically targeting the human SOD1 gene (SOD1). Injection of this vector into various muscle groups of mice engineered to overexpress a mutated form of human SOD1 (SOD1(G93A)) resulted in an efficient and specific reduction of SOD1 expression and improved survival of vulnerable motor neurons in the brainstem and spinal cord. Furthermore, SOD1 silencing mediated an improved motor performance in these animals, resulting in a considerable delay in the onset of ALS symptoms by more than 100% and an extension in survival by nearly 80% of their normal life span. These data are the first to show a substantial extension of survival in an animal model of a fatal, dominantly inherited neurodegenerative condition using RNAi and provide the highest therapeutic efficacy observed in this field to date.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Degeneración Nerviosa , Interferencia de ARN , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/terapia , Animales , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Humanos , Lentivirus/genética , Ratones , Ratones Transgénicos , Mutación , ARN Interferente Pequeño , Tasa de Supervivencia , Transfección
11.
J Clin Invest ; 114(12): 1726-31, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15599397

RESUMEN

Spinal muscular atrophy (SMA) is a frequent recessive autosomal disorder. It is caused by mutations or deletion of the telomeric copy of the survival motor neuron (SMN) gene, leading to depletion in SMN protein levels. The treatment rationale for SMA is to halt or delay the degeneration of motor neurons, but to date there are no effective drug treatments for this disease. We have previously demonstrated that pseudotyping of the nonprimate equine infectious anemia virus (using the lentivector gene transfer system) with the glycoprotein of the Evelyn-Rokitnicki-Abelseth strain of the rabies virus confers retrograde axonal transport on these vectors. Here, we report that lentivector expressing human SMN was successfully used to restore SMN protein levels in SMA type 1 fibroblasts. Multiple single injections of a lentiviral vector expressing SMN in various muscles of SMA mice restored SMN to motor neurons, reduced motor neuron death, and increased the life expectancy by an average of 3 and 5 days (20% and 38%) compared with LacZ and untreated animals, respectively. Further extension of survival by SMN expression constructs will likely require a knowledge of when and/or where high levels of SMN are needed.


Asunto(s)
Lentivirus/genética , Proteínas del Tejido Nervioso/genética , Animales , Línea Celular , Núcleo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos , Humanos , Inmunohistoquímica , Operón Lac , Ratones , Microscopía Fluorescente , Neuronas Motoras/metabolismo , Proteínas de Unión al ARN , Proteínas del Complejo SMN , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA