Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Sci Adv ; 10(10): eadl1122, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38446892

RESUMEN

Coxsackievirus B (CVB) infection of pancreatic ß cells is associated with ß cell autoimmunity and type 1 diabetes. We investigated how CVB affects human ß cells and anti-CVB T cell responses. ß cells were efficiently infected by CVB in vitro, down-regulated human leukocyte antigen (HLA) class I, and presented few, selected HLA-bound viral peptides. Circulating CD8+ T cells from CVB-seropositive individuals recognized a fraction of these peptides; only another subfraction was targeted by effector/memory T cells that expressed exhaustion marker PD-1. T cells recognizing a CVB epitope cross-reacted with ß cell antigen GAD. Infected ß cells, which formed filopodia to propagate infection, were more efficiently killed by CVB than by CVB-reactive T cells. Our in vitro and ex vivo data highlight limited CD8+ T cell responses to CVB, supporting the rationale for CVB vaccination trials for type 1 diabetes prevention. CD8+ T cells recognizing structural and nonstructural CVB epitopes provide biomarkers to differentially follow response to infection and vaccination.


Asunto(s)
Infecciones por Coxsackievirus , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Humanos , Linfocitos T CD8-positivos , Anticuerpos , Epítopos , Péptidos , Antivirales
2.
bioRxiv ; 2023 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-37662376

RESUMEN

Coxsackievirus B (CVB) infection of pancreatic ß cells is associated with ß-cell autoimmunity. We investigated how CVB impacts human ß cells and anti-CVB T-cell responses. ß cells were efficiently infected by CVB in vitro, downregulated HLA Class I and presented few, selected HLA-bound viral peptides. Circulating CD8+ T cells from CVB-seropositive individuals recognized only a fraction of these peptides, and only another sub-fraction was targeted by effector/memory T cells that expressed the exhaustion marker PD-1. T cells recognizing a CVB epitope cross-reacted with the ß-cell antigen GAD. Infected ß cells, which formed filopodia to propagate infection, were more efficiently killed by CVB than by CVB-reactive T cells. Thus, our in-vitro and ex-vivo data highlight limited T-cell responses to CVB, supporting the rationale for CVB vaccination trials for type 1 diabetes prevention. CD8+ T cells recognizing structural and non-structural CVB epitopes provide biomarkers to differentially follow response to infection and vaccination.

3.
PLoS Genet ; 16(11): e1009164, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33175861

RESUMEN

The chromosome translocations generating PAX3-FOXO1 and PAX7-FOXO1 chimeric proteins are the primary hallmarks of the paediatric fusion-positive alveolar subtype of Rhabdomyosarcoma (FP-RMS). Despite the ability of these transcription factors to remodel chromatin landscapes and promote the expression of tumour driver genes, they only inefficiently promote malignant transformation in vivo. The reason for this is unclear. To address this, we developed an in ovo model to follow the response of spinal cord progenitors to PAX-FOXO1s. Our data demonstrate that PAX-FOXO1s, but not wild-type PAX3 or PAX7, trigger the trans-differentiation of neural cells into FP-RMS-like cells with myogenic characteristics. In parallel, PAX-FOXO1s remodel the neural pseudo-stratified epithelium into a cohesive mesenchyme capable of tissue invasion. Surprisingly, expression of PAX-FOXO1s, similar to wild-type PAX3/7, reduce the levels of CDK-CYCLIN activity and increase the fraction of cells in G1. Introduction of CYCLIN D1 or MYCN overcomes this PAX-FOXO1-mediated cell cycle inhibition and promotes tumour growth. Together, our findings reveal a mechanism that can explain the apparent limited oncogenicity of PAX-FOXO1 fusion transcription factors. They are also consistent with certain clinical reports indicative of a neural origin of FP-RMS.


Asunto(s)
Transdiferenciación Celular/genética , Transformación Celular Neoplásica/genética , Proteínas de Fusión Oncogénica/metabolismo , Factores de Transcripción Paired Box/metabolismo , Rabdomiosarcoma Alveolar/genética , Animales , Biopsia , Embrión de Pollo , Niño , Ciclina D1/genética , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína Proto-Oncogénica N-Myc/genética , Invasividad Neoplásica/genética , Células-Madre Neurales/patología , Tubo Neural/citología , Proteínas de Fusión Oncogénica/genética , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Factores de Transcripción Paired Box/genética , Rabdomiosarcoma Alveolar/patología , Fase S/genética
4.
Expert Rev Vaccines ; 17(11): 977-987, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30365908

RESUMEN

INTRODUCTION: One of the essential mechanisms of virus infection control is cell-mediated cytotoxicity, which can act in an antibody-dependent or -independent fashion and is provided by different effector cells. The role of CD8 T-cells in infection control and in affecting the pathological outcome of different types of infection has been demonstrated in numerous animal studies. Despite this, their role in controlling human influenza infection is not fully understood. Especially, knowledge about their induction and turnover in human influenza infection is limited. Differences in the development of CD8 T-cells after influenza infection or immunizations should be explored in detail, in relation to the bioaccessibility of influenza antigens, site of application and distribution routes. AREAS COVERED: This review focuses on the basics of CD8 T-cell immune response both in human influenza infection and after administration of inactivated or live attenuated influenza vaccine. Some aspects of the accessibility, distribution and presentation of influenza antigens to CD8 T-cells are described. EXPERT COMMENTARY: The CD8 T-cell response is an essential connection between innate and antibody-mediated responses, which are all-important for influenza control. We hypothesize that immunization with live influenza vaccine is the most straightforward artificial way to induce an efficient influenza-specific CD8 T-cell response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/prevención & control , Animales , Formación de Anticuerpos/inmunología , Humanos , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Vacunación/métodos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
5.
Hum Vaccin Immunother ; 14(12): 2964-2970, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30024831

RESUMEN

The development of viral vector vaccines against various pathogens for which conventional vaccination approaches are not applicable has been a priority for a number of years. One promising approach is the insertion of immunodominant conservative cytotoxic T-cell (CTL) epitopes into the genome of a viral vector, which then delivers these epitopes to target cells, inducing immunity. Many different viruses have been assessed as viral vectors for CTL-based vaccines, but only a few of them are clinically relevant, mainly because of safety issues and limited knowledge about their performance in humans. In this regard, the use of licensed cold-adapted live attenuated influenza vaccine (LAIV) viruses as a vector delivery system has clear advantages for CTL-based vector vaccines against other respiratory pathogens: LAIV is known to induce all arms of the adaptive immune system and is administered via nasal spray, and its production process is relatively easy and inexpensive. Here we present the first results of the use of an LAIV backbone for designing a CTL epitope-based vaccine against respiratory syncytial virus (RSV). The chimeric LAIV-RSV vaccine candidates were attenuated in mice and induced strong, fully functional CTL immunity in this animal model.

6.
Infect Genet Evol ; 64: 95-104, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29929009

RESUMEN

Live attenuated influenza vaccines (LAIVs) are promising tools for the induction of broad protection from influenza due to their ability to stimulate cross-reactive T cells against influenza pathogens. One of the major targets for cytotoxic T-cell immunity is viral nucleoprotein (NP), which is relatively conserved among antigenically distant influenza viruses. Nevertheless, a diversity of epitope composition has been found in the NP protein of different lineages of influenza A viruses. The H2N2 master donor virus which is currently used as a backbone for the LAIV and donor of the six genomic segments encoding the internal proteins, A/Leningrad/134/17/57 (MDV Len/17), was isolated 60 years ago. As such, NP-specific T-cell immunity induced upon vaccination with classical LAIVs with a 6:2 genome composition containing this older NP might be suboptimal against currently circulating influenza viruses. In this study, a panel of H3N2 LAIV candidates with wild-type NP genes derived from circulating viruses were generated by reverse genetics (5:3 genome composition). These viruses displayed the cold adaptation and temperature sensitivity phenotypes of MDV Len/17 in vitro. LAIVs with both 6:2 and 5:3 genome compositions were attenuated and replicated to a similar extent in the upper respiratory tract of ferrets. LAIVs were immunogenic as high neutralizing and hemagglutination inhibition serum antibody titers were detected 21 days after infection. All vaccinated animals were protected against infection with heterologous H3N2 influenza A viruses. Thus, LAIV with a 5:3 genome composition is safe, immunogenic and can induce cross-protective immunity.


Asunto(s)
Enfermedades de los Animales/prevención & control , Inmunogenicidad Vacunal , Subtipo H3N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Nucleoproteínas/inmunología , Infecciones por Orthomyxoviridae/veterinaria , Vacunas Atenuadas/inmunología , Enfermedades de los Animales/inmunología , Enfermedades de los Animales/virología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Femenino , Hurones , Genoma Viral , Subtipo H3N2 del Virus de la Influenza A/genética , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/genética , Masculino , Pruebas de Neutralización , Nucleoproteínas/genética , Vacunación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética
7.
J Infect Dis ; 218(3): 406-417, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29746640

RESUMEN

Epidemiological studies have observed that the seasonal peak incidence of influenza virus infection is sometimes separate from the peak incidence of human respiratory syncytial virus (hRSV) infection, with the peak incidence of hRSV infection delayed. This is proposed to be due to viral interference, whereby infection with one virus prevents or delays infection with a different virus. We investigated viral interference between hRSV and 2009 pandemic influenza A(H1N1) virus (A[H1N1]pdm09) in the ferret model. Infection with A(H1N1)pdm09 prevented subsequent infection with hRSV. Infection with hRSV reduced morbidity attributed to infection with A(H1N1)pdm09 but not infection, even when an increased inoculum dose of hRSV was used. Notably, infection with A(H1N1)pdm09 induced higher levels of proinflammatory cytokines, chemokines, and immune mediators in the ferret than hRSV. Minimal cross-reactive serological responses or interferon γ-expressing cells were induced by either virus ≥14 days after infection. These data indicate that antigen-independent mechanisms may drive viral interference between unrelated respiratory viruses that can limit subsequent infection or disease.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Infecciones por Orthomyxoviridae/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/crecimiento & desarrollo , Interferencia Viral , Animales , Anticuerpos Antivirales , Modelos Animales de Enfermedad , Hurones , Inmunidad Celular , Inmunidad Humoral , Interferón gamma/análisis , Leucocitos Mononucleares/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Virus Sincitial Respiratorio/patología , Análisis de Supervivencia
8.
Virology ; 518: 313-323, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29574336

RESUMEN

The development of influenza vaccines that can provide broad protection against all drifted seasonal virus variants, zoonotic infections and emerging pandemic strains, has been a priority for two decades. Here we propose a strategy of inducing broadly-reactive anti-stalk antibody by sequential immunizations with live attenuated influenza vaccines (LAIVs) expressing chimeric HAs (cHAs). These vaccines are designed to contain identical hemagglutinin stalk domains from H1N1 virus but antigenically unrelated globular head domains from avian influenza virus subtypes H5, H8 and H9. Mouse experiments demonstrated enhanced cross-protection of cHA-containing LAIVs compared to the relevant vaccine viruses expressing natural HAs, and this enhanced protection was driven by stalk-HA-reactive IgG antibodies. The establishment of fully functional cross-protective immunity after two doses of cHA LAIV vaccination in naïve animals suggests that a similar effect might be expected after a single cHA LAIV dose in primed individuals, or after two to three doses in naïve children.


Asunto(s)
Anticuerpos Antivirales/sangre , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/inmunología , Animales , Protección Cruzada , Modelos Animales de Enfermedad , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Inmunoglobulina G/sangre , Vacunas contra la Influenza/administración & dosificación , Ratones , Infecciones por Orthomyxoviridae/prevención & control , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
9.
J Virol ; 92(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29187546

RESUMEN

Small-animal models have been used to obtain many insights regarding the pathogenesis and immune responses induced following infection with human respiratory syncytial virus (hRSV). Among those described to date, infections in cotton rats, mice, guinea pigs, chinchillas, and Syrian hamsters with hRSV strains Long and/or A2 have been well characterized, although clinical isolates have also been examined. Ferrets are also susceptible to hRSV infection, but the pathogenesis and immune responses elicited following infection have not been well characterized. Here, we describe the infection of adult ferrets with hRSV Long or A2 via the intranasal route and characterized virus replication, as well as cytokine induction, in the upper and lower airways. Virus replication and cytokine induction during the acute phase of infection (days 0 to 15 postinfection) were similar between the two strains, and both elicited high levels of F glycoprotein-specific binding and neutralizing antibodies following virus clearance (days 16 to 22 postinfection). Importantly, we demonstrate transmission from experimentally infected donor ferrets to cohoused naive recipients and have characterized virus replication and cytokine induction in the upper airways of infected contact animals. Together, these studies provide a direct comparison of the pathogenesis of hRSV Long and A2 in ferrets and highlight the potential of this animal model to study serological responses and examine interventions that limit transmission of hRSV.IMPORTANCE Ferrets have been widely used to study pathogenesis, immunity, and transmission following human influenza virus infections; however, far less is known regarding the utility of the ferret model to study hRSV infections. Following intranasal infection of adult ferrets with the well-characterized Long or A2 strain of hRSV, we report virus replication and cytokine induction in the upper and lower airways, as well as the development of virus-specific humoral responses. Importantly, we demonstrate transmission of hRSV from experimentally infected donor ferrets to cohoused naive recipients. Together, these findings significantly enhance our understanding of the utility of the ferret as a small-animal model to investigate aspects of hRSV pathogenesis and immunity.


Asunto(s)
Modelos Animales de Enfermedad , Inmunidad Humoral/inmunología , Infecciones por Virus Sincitial Respiratorio/transmisión , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/patogenicidad , Infecciones del Sistema Respiratorio/virología , Animales , Hurones , Células HeLa , Humanos , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitial Respiratorio Humano/inmunología , Infecciones del Sistema Respiratorio/inmunología , Carga Viral , Replicación Viral
10.
Biomed Res Int ; 2017: 9359276, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28210631

RESUMEN

Since conserved viral proteins of influenza virus, such as nucleoprotein (NP) and matrix 1 protein, are the main targets for virus-specific CD8+ cytotoxic T-lymphocytes (CTLs), we hypothesized that introduction of the NP gene of wild-type virus into the genome of vaccine reassortants could lead to better immunogenicity and afford better protection. This paper describes in vitro and in vivo preclinical studies of two new reassortants of pandemic H1N1 live attenuated influenza vaccine (LAIV) candidates. One had the hemagglutinin (HA) and neuraminidase (NA) genes from A/South Africa/3626/2013 H1N1 wild-type virus on the A/Leningrad/134/17/57 master donor virus backbone (6 : 2 formulation) while the second had the HA, NA, and NP genes of the wild-type virus on the same backbone (5 : 3 formulation). Although both LAIVs induced similar antibody immune responses, the 5 : 3 LAIV provoked greater production of virus-specific CTLs than the 6 : 2 variant. Furthermore, the 5 : 3 LAIV-induced CTLs had higher in vivo cytotoxic activity, compared to 6 : 2 LAIV. Finally, the 5 : 3 LAIV candidate afforded greater protection against infection and severe illness than the 6 : 2 LAIV. Inclusion in LAIV of the NP gene from wild-type influenza virus is a new approach to inducing cross-reactive cell-mediated immune responses and cross protection against pandemic influenza.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/clasificación , Vacunas contra la Influenza/inmunología , Neuraminidasa/inmunología , Nucleoproteínas/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Protección Cruzada , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/uso terapéutico , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/uso terapéutico , Ratones , Neuraminidasa/genética , Neuraminidasa/uso terapéutico , Nucleoproteínas/genética , Nucleoproteínas/uso terapéutico , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/uso terapéutico
11.
Virology ; 500: 209-217, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27829176

RESUMEN

This study sought to improve an existing live attenuated influenza vaccine (LAIV) by including nucleoprotein (NP) from wild-type virus rather than master donor virus (MDV). H7N9 LAIV reassortants with 6:2 (NP from MDV) and 5:3 (NP from wild-type virus) genome compositions were compared with regard to their growth characteristics, induction of humoral and cellular immune responses in mice, and ability to protect mice against homologous and heterologous challenge viruses. Although, in general, the 6:2 reassortant induced greater cell-mediated immunity in C57BL6 mice than the 5:3 vaccine, mice immunized with the 5:3 LAIV were better protected against heterologous challenge. The 5:3 LAIV-induced CTLs also had better in vivo killing activity against target cells loaded with the NP366 epitope of recent influenza viruses. Modification of the genome of reassortant vaccine viruses by incorporating the NP gene from wild-type viruses represents a simple strategy to improve the immunogenicity and cross-protection of influenza vaccines.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/inmunología , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Nucleoproteínas/inmunología , Vacunas Atenuadas/inmunología , Animales , Anticuerpos Antivirales/inmunología , Frío , Protección Cruzada , Femenino , Humanos , Inmunidad Celular , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/fisiología , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/prevención & control , Gripe Humana/virología , Ratones , Ratones Endogámicos C57BL , Nucleoproteínas/administración & dosificación , Nucleoproteínas/genética , Virus Reordenados/inmunología , Virus Reordenados/patogenicidad , Virus Reordenados/fisiología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Virulencia
13.
Lancet Infect Dis ; 16(3): 303-10, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26673391

RESUMEN

BACKGROUND: H7N9 avian influenza viruses characterised by high virulence and presence of mammalian adaptation markers have pandemic potential. Specific influenza vaccines remain the main defence. We assessed the safety and immunogenicity of an H7N9 live attenuated influenza vaccine (LAIV) candidate in healthy adult volunteers. METHODS: We did a phase 1, double-blind, randomised, placebo-controlled trial in Saint Petersburg, Russia. Eligible participants were healthy adults aged 18-49 years. The participants were randomised 3:1 to receive live vaccine or placebo, according to a computer-generated randomisation scheme. Two doses of vaccine or placebo were administered intranasally 28 days apart, each followed by 7 day stays in hospital. Immune responses were assessed in nasal swabs, saliva, and serum specimens collected before and 28 days after each vaccine dose. The primary outcome was the safety profile. This trial is registered with ClinicalTrials.gov, number NCT02480101. FINDINGS: Between Oct 21, 2014, and Oct 31, 2014, 40 adults were randomised, of whom 39 (98%) were included in the per-protocol analysis (29 in the vaccine group and ten in the placebo group). The frequency of adverse events did not differ between the vaccine and placebo groups. Seroconversion of neutralising antibodies was seen in 14 participants after the first vaccine dose (48%, 95% CI 29·4-67·5) and 21 after the second vaccine dose (72%, 52·8-87·3). Immune responses were seen in 27 of 29 recipients (93%, 95% CI 77·2-99·2). Adverse effects were seen in 19 (63%) vaccine recipients and nine (90%) placebo recipients after the first dose and in nine (31%) and four (40%), respectively, after the second dose. These effects were mainly local and all were mild. INTERPRETATION: The H7N9 LAIV was well tolerated and safe and showed good immunogenicity. FUNDING: WHO.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza , Gripe Humana/prevención & control , Adulto , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/sangre , Método Doble Ciego , Femenino , Humanos , Esquemas de Inmunización , Gripe Humana/virología , Masculino , Persona de Mediana Edad , Vacunas Atenuadas , Adulto Joven
14.
Transl Res ; 170: 112-123, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25530473

RESUMEN

Platelet- and endothelial-derived microparticles influence the phenotype of peripheral blood leukocytes and induce production of proinflammatory cytokines. The influence of blood plasma microparticles of pregnant women on the surface receptor expression on intact or activated monocytes is still unexplored. This study was carried out to test the hypothesis that peripheral blood microparticles of women with normal pregnancy and women with preeclampsia have different influence on the expression of surface molecules on monocytes. The objective of the study was to evaluate the influence of blood plasma microparticles of pregnant women on the phenotypic properties of intact and activated THP-1 monocytes. Microparticles were isolated from peripheral blood samples of nonpregnant women, healthy pregnant women, and women with preeclampsia. THP-1 cell line was used as a model of monocytes. Microparticles of nonpregnant women decreased CD18, CD49d, and CD54 expressions and increased CD11c, CD31, CD47, and vascular endothelial growth factor receptor 2 expressions. Microparticles of healthy pregnant women increased CD18, CD54, and integrin ß7 expressions and decreased CD11a and CD29 expressions. Microparticles of women with preeclampsia decreased CD18 expression on tumor necrosis factor α (TNF-α)-activated ТНР-1 cells. Microparticles of nonpregnant women, women with normal pregnancy, and pregnant women with preeclampsia decreased CD181 expression on intact and TNF-α-activated THP-1 cells. Therefore, blood plasma microparticles of women with normal pregnancy and women with preeclampsia have different influences on the expression of surface molecules on THP-1 monocytes.


Asunto(s)
Antígenos CD/metabolismo , Micropartículas Derivadas de Células , Monocitos/fisiología , Preeclampsia/sangre , Adulto , Línea Celular , Micropartículas Derivadas de Células/ultraestructura , Femenino , Humanos , Microscopía de Fuerza Atómica , Embarazo , Factor de Necrosis Tumoral alfa/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Hum Vaccin Immunother ; 11(12): 2839-48, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26697973

RESUMEN

During the past decade, a number of H5 subtype influenza vaccines have been developed and tested in clinical trials, but most of them induced poor serum antibody responses prompting the evaluation of novel vaccination approaches. One of the most promising ones is a "prime-boost" strategy, which could result in the induction of prompt and robust immune responses to a booster influenza vaccine following priming with homologous or heterologous vaccine strains. In our study we evaluated immunogenicity of an adjuvanted A(H5N1) inactivated influenza vaccine (IIV) in healthy adult subjects who received A(H5N2) live attenuated influenza vaccine (LAIV) 1.5 years earlier and compared this with a group of naïve subjects. We found that priming with A(H5N2) LAIV induced a long-lasting B-cell immunological memory against influenza A(H5N1) virus, which was brought on by more prompt and vigorous antibody production to a single dose of A(H5N1) IIV in the primed group, compared to the naïve controls. Thus, by day 28 after the first booster dose, the hemagglutination inhibition and neutralizing (MN) antibody titer rises were 17.2 and 30.8 in the primed group, compared to 2.3 and 8.0 in the control group, respectively. The majority (79%) of the primed individuals achieved seroprotective MN antibody titers at 7 days after the first dose of the IIV. All LAIV-primed volunteers had MN titers ≥ 1:40 by Day 28 after one dose of IIV, whereas only 58% subjects from the naïve control group developed similar immune responses at this time point. The second A(H5N1) IIV dose did not increase the immune response in the LAIV-primed group, whereas 2 doses of IIV were required for naïve volunteers to develop significant immune responses. These findings were of special significance since Russian-based LAIV technology has been licensed to WHO, through whom the vaccine has been provided to vaccine manufacturers in India, China and Thailand - countries particularly vulnerable to a pandemic influenza. The results of our study will be useful to inform the development of vaccination strategies in these countries in the event of a pandemic.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/inmunología , Adulto , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Reacciones Cruzadas/inmunología , Femenino , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunización Secundaria , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Memoria Inmunológica/inmunología , Masculino , Vacunación
16.
Influenza Other Respir Viruses ; 6(2): 119-26, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21819548

RESUMEN

PURPOSE: The main purpose of vaccination is to generate immunological memory providing enhanced immune responses against infectious pathogens. The standard and most commonly used assay for influenza vaccine immunogenicity evaluation is a hemagglutination inhibition assay (HAI). It is clear now that HAI assay is unable to properly assess the proven protective immunity elicited by live attenuated influenza vaccines (LAIV). New methods need to be developed for more accurate LAIV immunogenicity assessment and prediction of vaccine efficacy among target populations. OBJECTIVE: Randomized placebo-controlled study of memory B- and T-cell responses to intranasal LAIV in young adults. METHODS: A total of 56 healthy young adults 18-20 years old received seasonal monovalent LAIV. Mucosal memory B-cell responses were measured by IgA avidity assessment in nasal swabs. CD4 memory T cells in peripheral blood were examined by the expression of CD45RO marker and in functional test by the ability of virus-specific T cells to maintain the trogocytosis with antigen-loaded target cells. RESULTS: Intranasal LAIV immunization enhances mucosal IgA avidity even without reliable increases in antibody titers. At the day 21 after vaccination, up to 40% of subjects demonstrated significant increases in both total and virus-specific CD4 memory T cells that were observed regardless of seroconversion rate measured by HAI assay. CONCLUSION: The data suggest that immunogenicity of LAIV vaccines should be evaluated on the mucosal and cellular immunity basis. The assays applied could be used to support influenza clinical trials through preliminary screening of volunteers and subsequent measurement of anti-influenza in immunity.


Asunto(s)
Presentación de Antígeno , Linfocitos B/inmunología , Memoria Inmunológica , Vacunas contra la Influenza/inmunología , Orthomyxoviridae/inmunología , Virus Reordenados/inmunología , Linfocitos T/inmunología , Administración Intranasal , Adolescente , Anticuerpos Antivirales/inmunología , Afinidad de Anticuerpos , Antígenos CD4/análisis , Experimentación Humana , Humanos , Inmunidad Mucosa , Inmunoglobulina A/inmunología , Vacunas contra la Influenza/administración & dosificación , Antígenos Comunes de Leucocito/análisis , Placebos/administración & dosificación , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Adulto Joven
17.
Vaccine ; 27(19): 2580-7, 2009 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-19428864

RESUMEN

The first and most significant barrier against influenza infection is the mucosal-associated lymphoid tissue of the upper airways and rodent nasopharyngeal-associated lymphoid tissue (NALT) is considered equivalent to the lymphoid tissue of human Valdryer's ring. This study is the first attempt to analyze and compare local and systemic cellular and antibody immune responses in NALT and spleen in a mouse model of experimental influenza infection and intranasal vaccination with LAIV (live attenuated reassortant influenza vaccine). It was shown that the vaccine strain completely inherited the ability to induce high-grade local antibody responses (secretory IgA+IgG+IgM), local cellular lymphoproliferative activity, CD4(+), CD8(+) and CD19(+) lymphocyte and cytokine production responses from the virulent parental strain but it had less capacity to stimulate production of serum IgG, accumulation of CD8(+) cells and IFN-gamma production in the spleen. Primary non-complicated influenza infection and primary vaccination were accompanied by a short-term (24h) increase in the levels of lymphocyte apoptosis in both NALT and spleen. However, experimental data indicated that vaccination with LAIV and uncomplicated forms of influenza infection did not influence immune system apoptosis following a secondary immune response.


Asunto(s)
Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Gripe Humana/patología , Tejido Linfoide/inmunología , Mucosa Respiratoria/inmunología , Bazo/inmunología , Animales , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/sangre , Apoptosis , Proliferación Celular , Femenino , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos CBA , Subgrupos de Linfocitos T/inmunología , Vacunas Atenuadas/inmunología
18.
Influenza Other Respir Viruses ; 2(5): 165-74, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19453421

RESUMEN

BACKGROUND: Asthmatics in particular have a need for influenza vaccines because influenza infection is a frequent cause of hospitalization of patients with bronchial asthma. Currently, only inactivated influenza vaccines are recommended for influenza prevention in asthma sufferers. OBJECTIVE: The aim of our study was to analyze and compare the effects of influenza infection and vaccination with live attenuated influenza vaccine (LAIV) on different phases of experimental murine allergic bronchial asthma (acute asthma and remission phase) and on subsequent exposure to allergen in sensitized animals. METHODS: Ovalbumin (OVA)-specific serum IgE levels, IL-4 production by spleen and lung lymphocytes, and histological changes in the lungs of mice infected with pathogenic virus or LAIV were studied at two phases of OVA-induced bronchial asthma (acute asthma and remission). Results Infection with pathogenic virus both in acute asthma and remission led to asthma exacerbation associated with the production of OVA-specific IgE, IL-4 and significant inflammatory infiltration in airways. Infection, even after complete virus clearance, induced the aggravation of lung inflammation and IgE production in asthmatic mice additionally exposed to OVA. Immunization with LAIV at remission did not enhance allergic inflammatory changes in the lung, OVA-specific IgE or IL-4 production. Then after additional OVA exposure, histological and immunological changes in these mice were the same as in the control group. CONCLUSIONS: Influenza infection provokes asthma exacerbation regardless of the disease phase. Immunization with LAIV during the remission phase of bronchial asthma is safe and does not interfere upon subsequent contact of asthma sufferers with allergen.


Asunto(s)
Asma/inmunología , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulina E/sangre , Interleucina-4/metabolismo , Pulmón/inmunología , Pulmón/patología , Ratones , Bazo/inmunología , Linfocitos T/inmunología , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...