Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Commun Biol ; 6(1): 102, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36702861

RESUMEN

Protein acetylation plays a key role in regulating cellular processes and is subject to aberrant control in diverse pathologies. Although histone deacetylase (HDAC) inhibitors are approved drugs for certain cancers, it is not known whether they can be deployed in other therapeutic contexts. We have explored the clinical HDAC inhibitor, zabadinostat/CXD101, and found that it is a stand-alone regulator of the adaptive immune response. Zabadinostat treatment increased expression of MHC class I and II genes in a variety of cells, including dendritic cells (DCs) and healthy tissue. Remarkably, zabadinostat enhanced the activity of DCs, and CD4 and CD8 T lymphocytes. Using an antigenic peptide presented to the immune system by MHC class I, zabadinostat caused an increase in antigen-specific CD8 T lymphocytes. Further, mice immunised with covid19 spike protein and treated with zabadinostat exhibit enhanced covid19 neutralising antibodies and an increased level of T lymphocytes. The enhanced humoral response reflected increased activity of T follicular helper (Tfh) cells and germinal centre (GC) B cells. Our results argue strongly that zabadinostat has potential to augment diverse therapeutic agents that act through the immune system.


Asunto(s)
COVID-19 , Inmunidad Humoral , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Inhibidores de Histona Desacetilasas/farmacología , Inmunidad Adaptativa , Antígenos
2.
Nat Commun ; 14(1): 1078, 2023 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-36841868

RESUMEN

Protein arginine methyltransferase (PRMT) 5 is over-expressed in a variety of cancers and the master transcription regulator E2F1 is an important methylation target. We have explored the role of PRMT5 and E2F1 in regulating the non-coding genome and report here a striking effect on long non-coding (lnc) RNA gene expression. Moreover, many MHC class I protein-associated peptides were derived from small open reading frames in the lncRNA genes. Pharmacological inhibition of PRMT5 or adjusting E2F1 levels qualitatively altered the repertoire of lncRNA-derived peptide antigens displayed by tumour cells. When presented to the immune system as either ex vivo-loaded dendritic cells or expressed from a viral vector, lncRNA-derived peptides drove a potent antigen-specific CD8 T lymphocyte response, which translated into a significant delay in tumour growth. Thus, lncRNA genes encode immunogenic peptides that can be deployed as a cancer vaccine.


Asunto(s)
Neoplasias , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Neoplasias/genética , Neoplasias/terapia , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Péptidos/genética , Linfocitos T CD8-positivos , Proteína-Arginina N-Metiltransferasas
3.
JCI Insight ; 7(13)2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35608920

RESUMEN

The role of immune responses to previously seen endemic coronavirus epitopes in severe acute respiratory coronavirus 2 (SARS-CoV-2) infection and disease progression has not yet been determined. Here, we show that a key characteristic of fatal outcomes with coronavirus disease 2019 (COVID-19) is that the immune response to the SARS-CoV-2 spike protein is enriched for antibodies directed against epitopes shared with endemic beta-coronaviruses and has a lower proportion of antibodies targeting the more protective variable regions of the spike. The magnitude of antibody responses to the SARS-CoV-2 full-length spike protein, its domains and subunits, and the SARS-CoV-2 nucleocapsid also correlated strongly with responses to the endemic beta-coronavirus spike proteins in individuals admitted to an intensive care unit (ICU) with fatal COVID-19 outcomes, but not in individuals with nonfatal outcomes. This correlation was found to be due to the antibody response directed at the S2 subunit of the SARS-CoV-2 spike protein, which has the highest degree of conservation between the beta-coronavirus spike proteins. Intriguingly, antibody responses to the less cross-reactive SARS-CoV-2 nucleocapsid were not significantly different in individuals who were admitted to an ICU with fatal and nonfatal outcomes, suggesting an antibody profile in individuals with fatal outcomes consistent with an "original antigenic sin" type response.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Antivirales , Formación de Anticuerpos , Epítopos , Humanos , SARS-CoV-2
4.
Infect Immun ; 79(8): 3328-37, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21628524

RESUMEN

Convincing correlates of protective immunity against tuberculosis have been elusive. In BALB/c mice, intranasal immunization with a replication-deficient recombinant adenovirus expressing Mycobacterium tuberculosis antigen 85A (adenovirus-85A) induces protective lower respiratory tract immunity against pulmonary challenge with Mycobacterium tuberculosis, while intradermal immunization with adenovirus-85A does not. Here we report that intranasal immunization with adenovirus-85A induces expression of the chemokine receptor CXCR6 on lung CD8 T lymphocytes, which is maintained for at least 3 months. CXCR6-positive antigen-specific T cell numbers are increased among bronchoalveolar lavage-recoverable cells. Similarly, intranasal immunization with recombinant antigen 85A with adjuvant induces CXCR6 expression on lung CD4 cells in BALB/c and C57BL/6 mice, while a synthetic ESAT6(1-20) peptide with adjuvant induces CXCR6 expression in C57BL/6 mice. Parenteral immunization fails to do so. Upregulation of CXCR6 is accompanied by a transient elevation of serum CXCL16 after intranasal immunization, and lung cells cultured ex vivo from mice immunized intranasally show increased production of CXCL16. Administration of CXCL16 and cognate antigen intranasally to mice previously immunized parenterally increases the number of antigen-specific T lymphocytes in the bronchoalveolar lavage-recoverable population, which mediates inhibition of the early growth of Mycobacterium tuberculosis after challenge. We conclude that expression of CXCR6 on lung T lymphocytes is a correlate of local protective immunity against Mycobacterium tuberculosis after intranasal immunization and that CXCR6 and CXCL16 play an important role in the localization of T cells within lung tissue and the bronchoalveolar lavage-recoverable compartment.


Asunto(s)
Biomarcadores/análisis , Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Receptores CXCR/análisis , Vacunas contra la Tuberculosis/inmunología , Aciltransferasas/genética , Aciltransferasas/inmunología , Adenoviridae/genética , Administración Intranasal , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Líquido del Lavado Bronquioalveolar/citología , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/química , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores CXCR6 , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
5.
J Virol ; 84(16): 8241-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534854

RESUMEN

We previously showed that agonistic antibodies to CD40 could substitute for CD4 T-cell help and prevent reactivation of murine gammaherpesvirus 68 (MHV-68) in the lungs of major histocompatibility complex (MHC) class II(-/-) (CII(-/-)) mice, which are CD4 T cell deficient. Although CD8 T cells were required for this effect, no change in their activity was detected in vitro. A key question was whether anti-CD40 treatment (or CD4 T-cell help) changed the function of CD8 T cells or another cell type in vivo. To address this question, in the present study, we showed that adoptive transfer of CD8 T cells from virus-infected wild-type mice or anti-CD40-treated CII(-/-) mice caused a significant reduction in lung viral titers, in contrast to those from control CII(-/-) mice. Anti-CD40 treatment also greatly prolonged survival of infected CII(-/-) mice. This confirms that costimulatory signals cause a change in CD8 T cells enabling them to maintain effective long-term control of MHV-68. We investigated the nature of this change and found that expression of the inhibitory receptor PD-1 was significantly increased on CD8 T cells in the lungs of MHV-68-infected CII(-/-), CD40(-/-), or CD80/86(-/-) mice, compared with that in wild-type or CD28/CTLA4(-/-) mice, correlating with the level of viral reactivation. Furthermore, blocking PD-1-PD-L1 interactions significantly reduced viral reactivation in CD4 T-cell-deficient mice. In contrast, the absence of another inhibitory receptor, NKG2A, had no effect. These data suggest that CD4 T-cell help programs a change in CD8 T-cell function mediated by altered PD-1 expression, which enables effective long-term control of MHV-68.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antígeno B7-1/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Glicoproteínas de Membrana/metabolismo , Péptidos/metabolismo , Rhadinovirus/inmunología , Traslado Adoptivo , Animales , Antígeno B7-H1 , Antígenos CD40/antagonistas & inhibidores , Femenino , Antígenos de Histocompatibilidad Clase II/genética , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Muerte Celular Programada 1 , Unión Proteica , Análisis de Supervivencia
6.
Am J Pathol ; 176(2): 800-11, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20042666

RESUMEN

Secondary bacterial infections that follow infection with influenza virus result in considerable morbidity and mortality in young children, the elderly, and immunocompromised individuals and may also significantly increase mortality in normal healthy adults during influenza pandemics. We herein describe a mouse model for investigating the interaction between influenza virus and the bacterium Haemophilus influenzae. Sequential infection with sublethal doses of influenza and H. influenzae resulted in synergy between the two pathogens and caused mortality in immunocompetent adult wild-type mice. Lethality was dependent on the interval between administration of the bacteria and virus, and bacterial growth was prolonged in the lungs of dual-infected mice, although influenza virus titers were unaffected. Dual infection induced severe damage to the airway epithelium and confluent pneumonia, similar to that observed in victims of the 1918 global influenza pandemic. Increased bronchial epithelial cell death was observed as early as 1 day after bacterial inoculation in the dual-infected mice. Studies using knockout mice indicated that lethality occurs via a mechanism that is not dependent on Fas, CCR2, CXCR3, interleukin-6, tumor necrosis factor, or Toll-like receptor-4 and does not require T or B cells. This model suggests that infection with virulent strains of influenza may predispose even immunocompetent individuals to severe illness on secondary infection with H. influenzae by a mechanism that involves innate immunity, but does not require tumor necrosis factor, interleukin-6, or signaling via Toll-like receptor-4.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Haemophilus/mortalidad , Haemophilus influenzae/fisiología , Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/mortalidad , Inmunidad Adaptativa/fisiología , Animales , Células Cultivadas , Perros , Infecciones por Haemophilus/complicaciones , Infecciones por Haemophilus/patología , Infecciones por Haemophilus/virología , Humanos , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Sobreinfección/inmunología , Sobreinfección/mortalidad , Sobreinfección/patología , Sobreinfección/virología , Carga Viral
7.
J Immunol ; 182(11): 7178-89, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19454715

RESUMEN

Type 1 IFNs, innate cytokines with important effector and immunomodulatory properties, are rapidly induced in the acute phase of many virus infections; however, this is generally a transient response that is not sustained during virus persistence. To gain insight into mechanisms that can contribute to down-regulation of type 1 IFN production during virus persistence, we analyzed type 1 IFN production during acute and chronic lymphocytic choriomeningitis virus (LCMV) infection. High-level type 1 IFN production was transiently up-regulated in cells including plasmacytoid and conventional dendritic cells (DCs) following LCMV infection of mice, but LCMV persistence was associated with only low-level type 1 IFN production. Nonetheless, chronically infected mice were able to up-regulate type 1 IFN production in response to TLR3, 7, and 9 ligands, albeit less efficiently than uninfected mice. Splenic DC numbers in mice chronically infected with LCMV were decreased, and the remaining cells exhibited a reduced response to TLR stimulation. LCMV-infected cell lines efficiently up-regulated type 1 IFN production following TLR ligation and infection with a DNA virus, but exhibited a defect in type 1 IFN induction following infection with Sendai, an RNA virus. This block in type 1 IFN production by infected cells, together with abnormalities in DC numbers and functions, likely contribute to the low-level type 1 IFN production in mice chronically infected with LCMV. Impairment of type 1 IFN production may both promote virus persistence and impact on host immunocompetence. Understanding the mechanisms involved may assist in development of strategies for control of virus persistence and superinfection.


Asunto(s)
Interferón Tipo I/biosíntesis , Animales , Virus ADN/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Células Plasmáticas/inmunología , Virus ARN/inmunología , Bazo/citología , Receptores Toll-Like/inmunología
8.
Vaccines (Basel) ; 8(2)2020 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-32295168

RESUMEN

Chronic hepatitis B virus (HBV) infection affects 257 million people globally. Current therapies suppress HBV but viral rebound occurs on cessation of therapy; novel therapeutic strategies are urgently required. To develop a therapeutic HBV vaccine that can induce high magnitude T cells to all major HBV antigens, we have developed a novel HBV vaccine using chimpanzee adenovirus (ChAd) and modified vaccinia Ankara (MVA) viral vectors encoding multiple HBV antigens. ChAd vaccine alone generated very high magnitude HBV specific T cell responses to all HBV major antigens. The inclusion of a shark Invariant (SIi) chain genetic adjuvant significantly enhanced the magnitude of T-cells against HBV antigens. Compared to ChAd alone vaccination, ChAd-prime followed by MVA-boost vaccination further enhanced the magnitude and breadth of the vaccine induced T cell response. Intra-cellular cytokine staining study showed that HBV specific CD8+ and CD4+ T cells were polyfunctional, producing combinations of IFNγ, TNF-α, and IL-2. In summary, we have generated genetically adjuvanted ChAd and MVA vectored HBV vaccines with the potential to induce high-magnitude T cell responses through a prime-boost therapeutic vaccination approach. These pre-clinical studies pave the way for new studies of HBV therapeutic vaccination in humans with chronic hepatitis B infection.

9.
J Virol ; 82(22): 11016-22, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18768977

RESUMEN

CD4 T cells are not essential for primary clearance of replicating murine gammaherpesvirus 68 (MHV-68) but are required for effective long-term control. The virus reactivates in the lungs of major histocompatibility complex class II-deficient (CII-/-) mice that lack functional CD4 T cells. CD40 ligand (CD40L) is upregulated on activated CD4 T cells, and it is thought that CD40-CD40L interactions are an important component of CD4 T-cell help. Our previous studies have shown that agonistic antibodies to CD40 can substitute for CD4 T-cell function in the long-term control of MHV-68. In the present study, we sought to identify the CD40-positive cell type mediating this effect. To address this question, we adoptively transferred MHV-68 peptide-pulsed CII(-/-) dendritic cells (DC) that had been treated with an agonistic antibody to CD40 into MHV-68-infected CII(-/-) recipients. Viral reactivation was significantly lower in mice injected with anti-CD40-treated DC than in those injected with control DC or in mice that did not receive any DC. However, in similar experiments with B cells, anti-CD40 treatment had no effect. We also investigated the requirement for CD40 expression on T cells by adoptive transfer of T cells from CD40(+/+) or CD40(-/-) mice into T-cell-deficient recipients that were subsequently infected with MHV-68. The results showed that CD40 expression on T cells is not necessary for preventing viral reactivation. Taken together, our data suggest that CD40 engagement on DC, but not on T or B cells, is essential for effective long-term control of MHV-68.


Asunto(s)
Linfocitos B/inmunología , Antígenos CD40/inmunología , Células Dendríticas/inmunología , Rhadinovirus/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antivirales/sangre , Femenino , Pulmón/virología , Ratones , Ratones Noqueados , Linfocitos T/inmunología , Ensayo de Placa Viral , Activación Viral/inmunología
10.
J Virol ; 82(23): 11970-5, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18818318

RESUMEN

CD4 T cells are dispensable for acute control of murine gammaherpesvirus 68 (MHV-68) but are necessary for effective long-term control of the virus by CD8 T cells. In contrast, protein kinase C theta (PKCtheta) is not essential for either acute or long-term viral control. However, we found that while either CD4 or CD8 T cells could mediate the clearance of MHV-68 from the lungs of PKCtheta(+/+) mice, PKCtheta(-/-) mice depleted of either subset failed to clear the virus. These data suggest that there are two alternative pathways for MHV-68 clearance, one dependent on CD4 T cells and the other on PKCtheta. Protection mediated by the latter appears to be short-lived. These observations may help to explain the differential requirement for PKCtheta in various models of CD8 T-cell activation and differences in the costimulatory requirements for acute and long-term viral control.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Isoenzimas/fisiología , Proteína Quinasa C/fisiología , Rhadinovirus/inmunología , Animales , Movimiento Celular , Citotoxicidad Inmunológica , Interferón gamma/biosíntesis , Pulmón/inmunología , Ratones , Proteína Quinasa C-theta
11.
Front Immunol ; 10: 197, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30894851

RESUMEN

Persistent virus infection can drive CD8+ T-cell responses which are markedly divergent in terms of frequency, phenotype, function, and distribution. On the one hand viruses such as Lymphocytic Choriomeningitis Virus (LCMV) Clone 13 can drive T-cell "exhaustion", associated with upregulation of checkpoint molecules, loss of effector functions, and diminished control of viral replication. On the other, low-level persistence of viruses such as Cytomegalovirus and Adenoviral vaccines can drive memory "inflation," associated with sustained populations of CD8+ T-cells over time, with maintained effector functions and a distinct phenotype. Underpinning these divergent memory pools are distinct transcriptional patterns-we aimed to compare these to explore the regulation of CD8+ T-cell memory against persistent viruses at the level of molecular networks and address whether dysregulation of specific modules may account for the phenotype observed. By exploring in parallel and also merging existing datasets derived from different investigators we attempted to develop a combined model of inflation vs. exhaustion and investigate the gene expression networks that are shared in these memory pools. In such comparisons, co-ordination of a critical module of genes driven by Tbx21 is markedly different between the two memory types. These exploratory data highlight both the molecular similarities as well as the differences between inflation and exhaustion and we hypothesize that co-ordinated regulation of a key genetic module may underpin the markedly different resultant functions and phenotypes in vivo-an idea which could be tested directly in future experiments.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interacciones Huésped-Patógeno/inmunología , Recuento de Linfocitos , Virosis/inmunología , Virosis/virología , Virus/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Interacciones Huésped-Patógeno/genética , Humanos , Memoria Inmunológica , Virosis/sangre , Virosis/genética
12.
Wellcome Open Res ; 4: 78, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31448339

RESUMEN

Background: Persistent viruses such as murine cytomegalovirus (MCMV) and adenovirus-based vaccines induce strong, sustained CD8 + T-cell responses, described as memory "inflation". These retain functionality, home to peripheral organs and are associated with a distinct transcriptional program. Methods: To further define the nature of the transcriptional mechanisms underpinning memory inflation at different sites we used single-cell RNA sequencing of tetramer-sorted cells from MCMV-infected mice, analyzing transcriptional networks in virus-specific populations in the spleen and gut intra-epithelial lymphocytes (IEL). Results: We provide a transcriptional map of T-cell memory and define a module of gene expression, which distinguishes memory inflation in spleen from resident memory T-cells (T RM) in the gut. Conclusions: These data indicate that CD8 + T-cell memory in the gut epithelium induced by persistent viruses and vaccines has a distinct quality from both conventional memory and "inflationary" memory which may be relevant to protection against mucosal infections.

13.
Cell Rep ; 23(3): 768-782, 2018 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669283

RESUMEN

The induction and maintenance of T cell memory is critical to the success of vaccines. A recently described subset of memory CD8+ T cells defined by intermediate expression of the chemokine receptor CX3CR1 was shown to have self-renewal, proliferative, and tissue-surveillance properties relevant to vaccine-induced memory. We tracked these cells when memory is sustained at high levels: memory inflation induced by cytomegalovirus (CMV) and adenovirus-vectored vaccines. In mice, both CMV and vaccine-induced inflationary T cells showed sustained high levels of CX3R1int cells exhibiting an effector-memory phenotype, characteristic of inflationary pools, in early memory. In humans, CX3CR1int CD8+ T cells were strongly induced following adenovirus-vectored vaccination for hepatitis C virus (HCV) (ChAd3-NSmut) and during natural CMV infection and were associated with a memory phenotype similar to that in mice. These data indicate that CX3CR1int cells form an important component of the memory pool in response to persistent viruses and vaccines in both mice and humans.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptor 1 de Quimiocinas CX3C/metabolismo , Citomegalovirus/inmunología , Memoria Inmunológica , Vacunas Virales/inmunología , Adenoviridae/genética , Adenoviridae/patogenicidad , Adulto , Animales , Linfocitos T CD8-positivos/inmunología , Receptor 1 de Quimiocinas CX3C/genética , Citomegalovirus/patogenicidad , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Hepacivirus/inmunología , Hepacivirus/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Vacunas Sintéticas/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo , Adulto Joven
15.
Cell Rep ; 13(8): 1578-88, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26586434

RESUMEN

Following exposure to vaccines, antigen-specific CD8(+) T cell responses develop as long-term memory pools. Vaccine strategies based on adenoviral vectors, e.g., those developed for HCV, are able to induce and sustain substantial CD8(+) T cell populations. How such populations evolve following vaccination remains to be defined at a transcriptional level. We addressed the transcriptional regulation of divergent CD8(+) T cell memory pools induced by an adenovector encoding a model antigen (beta-galactosidase). We observe transcriptional profiles that mimic those following infection with persistent pathogens, murine and human cytomegalovirus (CMV). Key transcriptional hallmarks include upregulation of homing receptors and anti-apoptotic pathways, driven by conserved networks of transcription factors, including T-bet. In humans, an adenovirus vaccine induced similar CMV-like phenotypes and transcription factor regulation. These data clarify the core features of CD8(+) T cell memory following vaccination with adenovectors and indicate a conserved pathway for memory development shared with persistent herpesviruses.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Vectores Genéticos/inmunología , Memoria Inmunológica/inmunología , Animales , Apoptosis/inmunología , Citomegalovirus/inmunología , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/inmunología , Vacunación/métodos
16.
PLoS One ; 6(11): e27477, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22110657

RESUMEN

BACKGROUND: BCG, the only licensed vaccine against tuberculosis, provides some protection against disseminated disease in infants but has little effect on prevention of adult pulmonary disease. Newer parenteral immunization prime boost regimes may provide improved protection in experimental animal models but are unproven in man so that there remains a need for new and improved immunization strategies. METHODS AND FINDINGS: Mice were immunized parenterally, intranasally or simultaneously by both routes with BCG or recombinant mycobacterial antigens plus appropriate adjuvants. They were challenged with Mycobacterium tuberculosis (Mtb) and the kinetics of Mtb growth in the lungs measured. We show that simultaneous immunization (SIM) of mice by the intranasal and parenteral routes is highly effective in increasing protection over parenteral BCG administration alone. Intranasal immunization induces local pulmonary immunity capable of inhibiting the growth of Mtb in the early phase (the first week) of infection, while parenteral immunization has a later effect on Mtb growth. Importantly, these two effects are additive and do not depend on priming and boosting the immune response. The best SIM regimes reduce lung Mtb load by up to 2 logs more than BCG given by either route alone. CONCLUSIONS: These data establish SIM as a novel and highly effective immunization strategy for Mtb that could be carried out at a single clinic visit. The efficacy of SIM does not depend on priming and boosting an immune response, but SIM is complementary to prime boost strategies and might be combined with them.


Asunto(s)
Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/inmunología , Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Inmunización/métodos , Tuberculosis/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Animales , Vías de Administración de Medicamentos , Femenino , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo , Tuberculosis/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
17.
Vaccine ; 28(32): 5179-84, 2010 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-20558252

RESUMEN

Intra-nasal administration of a recombinant adenovirus expressing Mycobacterium tuberculosis antigen 85A (Ad85A) has been shown to provide protection against challenge with M. tuberculosis. However the role of the upper respiratory tract associated lymphoid tissue, specifically the nasal associated lymphoid tissue (NALT), in providing protection has yet to be elucidated. Here we administered Ad85A to BALB/c mice alone or following BCG priming, using intranasal inocula targeting the whole respiratory tract or only the NALT, to show that Ad85A induces an immune response in the NALT insufficient to provide protection. Rather, Ad85A delivered through the respiratory tract must induce a deep lung immune response in order to protect against M. tuberculosis.


Asunto(s)
Aciltransferasas/inmunología , Antígenos Bacterianos/inmunología , Vacuna BCG/inmunología , Tejido Linfoide/inmunología , Nariz/inmunología , Tuberculosis/prevención & control , Adenoviridae/inmunología , Administración Intranasal , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Pulmón/inmunología , Ratones , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Vacunas Sintéticas/inmunología
18.
PLoS One ; 4(12): e8235, 2009 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20011050

RESUMEN

BACKGROUND: In pulmonary Mycobacterium tuberculosis (Mtb) infection, immune responses are delayed compared to other respiratory infections, so that antigen-specific cells are not detected in the lungs earlier than day 14. Even after parenteral immunization with Bacille Calmette Guerin (BCG) or a subunit vaccine, the immune response after Mtb challenge is only slightly accelerated and the kinetics of pulmonary Mtb growth do not differ between naïve and immunized animals up to day 14. METHODS AND FINDINGS: Mice were immunized intranasally with a recombinant adenovirus expressing mycobacterial antigen 85A (Ad85A), challenged by aerosol with Mtb and the kinetics of Mtb growth in the lungs measured. Intranasal immunization with Ad85A inhibits Mtb growth in the early phase of infection, up to day 8. Protection is sustained for at least 7 months and correlates with the presence of antigen-specific activated effector CD8 T cells in the lungs. Antigen 85A-specific T cells respond to antigen presenting cells from the lungs of mice immunized with Ad85A 23 weeks previously, demonstrating the persistence of antigen in the lungs. CONCLUSIONS/SIGNIFICANCE: Intranasal immunization with Ad85A can inhibit early growth of Mtb because it establishes a lung antigen depot and maintains an activated lung-resident lymphocyte population. We propose that an optimal immunization strategy for tuberculosis should aim to induce both lung and systemic immunity, targeting the early and late phases of Mtb growth.


Asunto(s)
Adenoviridae/inmunología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Vacunas contra la Tuberculosis/farmacología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/prevención & control , Vacunación , Adenoviridae/efectos de los fármacos , Administración Intranasal , Animales , Antígenos Bacterianos/inmunología , Citocinas/biosíntesis , Epítopos/inmunología , Femenino , Inmunidad/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Fenotipo , Factores de Tiempo , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/farmacología
19.
Eur J Immunol ; 36(8): 2150-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16856204

RESUMEN

CD45 is a leukocyte tyrosine phosphatase, essential for normal immune responses. We have studied the function of splenic dendritic cells of CD45(+/+), CD45(-/-), CD45RABC and CD45RO transgenic mice. We show that there are increased numbers of plasmacytoid dendritic cells in CD45(-/-) mice. DC of all mice are capable of responding to lymphocytic choriomeningitis virus (LCMV) infection by up-regulation of MHC and costimulatory molecules. DC of CD45(-/-) mice have an impaired capacity to produce type I interferons in response to LCMV infection in vivo. These data indicate that lack of CD45 expression in DC has a profound effect on their function. This is largely restored by CD45RABC or CD45RO transgenes.


Asunto(s)
Células Dendríticas/metabolismo , Interferón Tipo I/biosíntesis , Antígenos Comunes de Leucocito/metabolismo , Animales , Proliferación Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Antígenos Comunes de Leucocito/genética , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones , Ratones Transgénicos , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA