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










Base de datos
Intervalo de año de publicación
1.
Drug Metab Dispos ; 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37884391

RESUMEN

A STING (stimulator of interferon genes) agonist GSK3996915 under investigation in early discovery for hepatitis B was orally dosed to a mouse model for understanding the parent drug distribution in liver, the target organ. MALDI imaging mass spectrometry (IMS) was used to quantify the distribution of GSK3996915 in liver collected from mice administered a single oral dose at 90 mg/kg. GSK3996915 was detected with a zonal distribution localized in the portal triad and highly concentrated in the main bile ducts, indicating clearance through biliary excretion. High spatial resolution imaging showed the distribution of the parent drug localized to the cellular populations in the sinusoids including the Kupffer cells. Additionally, a series of drug-related metabolites were observed to be localized in the central zones of the liver. These results exemplify the potential of utilizing MALDI IMS for measuring not only quantitative drug distribution and target exposure, but also drug metabolism and elimination in a single suite of experiments. Significance Statement An integrated imaging approach utilizing MALDI IMS, immunohistochemistry (IHC), and histology was used to measure MALDI IMS complemented with other imaging techniques such as immunohistochemistry addressed the question of target exposure at the cellular level. Localized quantification of the parent drug in the target organ and identificaitonidentification of potential metabolites in the context of tissue histology were also achieved in one experimental suite to support characterization of pharmacokinetic properties of the drug in the early discovery stage.

2.
J Immunol ; 203(7): 1897-1908, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31484735

RESUMEN

Sepsis is characterized as life-threatening organ dysfunction caused by a dysregulated host immune response to infection. The purpose of this investigation was to determine the differential effect of sepsis on innate versus adaptive immunity, in humans, by examining RNA expression in specific immune cell subsets, including monocytes/macrophages and CD4 and CD8 T cells. A second aim was to determine immunosuppressive mechanisms operative in sepsis that might be amenable to immunotherapy. Finally, we examined RNA expression in peripheral cells from critically ill nonseptic patients and from cancer patients to compare the unique immune response in these disorders with that occurring in sepsis. Monocytes, CD4 T cells, and CD8 T cells from septic patients, critically ill nonseptic patients, patients with metastatic colon cancer, and healthy controls were analyzed by RNA sequencing. Sepsis induced a marked phenotypic shift toward downregulation of multiple immune response pathways in monocytes suggesting that impaired innate immunity may be fundamental to the immunosuppression that characterizes the disorder. In the sepsis cohort, there was a much more pronounced effect on gene transcription in CD4 T cells than in CD8 T cells. Potential mediators of sepsis-induced immunosuppression included Arg-1, SOCS-1, and SOCS-3, which were highly upregulated in multiple cell types. Multiple negative costimulatory molecules, including TIGIT, Lag-3, PD-1, and CTLA-4, were also highly upregulated in sepsis. Although cancer had much more profound effects on gene transcription in CD8 T cells, common immunosuppressive mechanisms were present in all disorders, suggesting that immunoadjuvant therapies that are effective in one disease may also be efficacious in the others.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Monocitos/inmunología , Neoplasias/inmunología , ARN Neoplásico/inmunología , Sepsis/inmunología , Análisis de Secuencia de ARN , Adulto , Anciano , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Enfermedad Crítica , Femenino , Humanos , Tolerancia Inmunológica , Masculino , Persona de Mediana Edad , Monocitos/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias/genética , Neoplasias/patología , Estudios Prospectivos , ARN Neoplásico/genética , Sepsis/genética , Sepsis/patología
4.
Nature ; 564(7736): 439-443, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30405246

RESUMEN

Stimulator of interferon genes (STING) is a receptor in the endoplasmic reticulum that propagates innate immune sensing of cytosolic pathogen-derived and self DNA1. The development of compounds that modulate STING has recently been the focus of intense research for the treatment of cancer and infectious diseases and as vaccine adjuvants2. To our knowledge, current efforts are focused on the development of modified cyclic dinucleotides that mimic the endogenous STING ligand cGAMP; these have progressed into clinical trials in patients with solid accessible tumours amenable to intratumoral delivery3. Here we report the discovery of a small molecule STING agonist that is not a cyclic dinucleotide and is systemically efficacious for treating tumours in mice. We developed a linking strategy to synergize the effect of two symmetry-related amidobenzimidazole (ABZI)-based compounds to create linked ABZIs (diABZIs) with enhanced binding to STING and cellular function. Intravenous administration of a diABZI STING agonist to immunocompetent mice with established syngeneic colon tumours elicited strong anti-tumour activity, with complete and lasting regression of tumours. Our findings represent a milestone in the rapidly growing field of immune-modifying cancer therapies.


Asunto(s)
Bencimidazoles/química , Bencimidazoles/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Diseño de Fármacos , Proteínas de la Membrana/agonistas , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/uso terapéutico , Humanos , Ligandos , Proteínas de la Membrana/inmunología , Ratones , Modelos Moleculares , Nucleótidos Cíclicos/metabolismo
5.
Cell Rep ; 18(8): 1906-1916, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28228257

RESUMEN

Long-lived plasma cells (PCs) in the bone marrow (BM) are a critical source of antibodies after infection or vaccination, but questions remain about the factors that control PCs. We found that systemic infection alters the BM, greatly reducing PCs and regulatory T (Treg) cells, a population that contributes to immune privilege in the BM. The use of intravital imaging revealed that BM Treg cells display a distinct behavior characterized by sustained co-localization with PCs and CD11c-YFP+ cells. Gene expression profiling indicated that BM Treg cells express high levels of Treg effector molecules, and CTLA-4 deletion in these cells resulted in elevated PCs. Furthermore, preservation of Treg cells during systemic infection prevents PC loss, while Treg cell depletion in uninfected mice reduced PC populations. These studies suggest a role for Treg cells in PC biology and provide a potential target for the modulation of PCs during vaccine-induced humoral responses or autoimmunity.


Asunto(s)
Células de la Médula Ósea/inmunología , Médula Ósea/inmunología , Células Plasmáticas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad/inmunología , Antígeno CTLA-4/inmunología , Inmunidad Humoral , Inmunofenotipificación/métodos , Ratones , Ratones Endogámicos C57BL
6.
Trends Mol Med ; 23(2): 165-180, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28109721

RESUMEN

The inflammasome is a cytoplasmic protein complex that processes interleukins (IL)-1ß and IL-18, and drives a form of cell death known as pyroptosis. Oligomerization of this complex is actually the second step of activation, and a priming step must occur first. This involves transcriptional upregulation of pro-IL-1ß, inflammasome sensor NLRP3, or the non-canonical inflammasome sensor caspase-11. An additional aspect of priming is the post-translational modification of particular inflammasome constituents. Priming is typically accomplished in vitro using a microbial Toll-like receptor (TLR) ligand. However, it is now clear that inflammasomes are activated during the progression of sterile inflammatory diseases such as atherosclerosis, metabolic disease, and neuroinflammatory disorders. Therefore, it is time to consider the endogenous factors and mechanisms that may prime the inflammasome in these conditions.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Enfermedad de Alzheimer/inmunología , Animales , Aterosclerosis/inmunología , Enfermedades Autoinflamatorias Hereditarias/inmunología , Humanos , Inmunidad Innata , Interleucina-1beta/inmunología , Enfermedades Metabólicas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Neuroinmunomodulación , Obesidad/inmunología , Receptores Toll-Like/inmunología
7.
J Immunol ; 194(8): 3784-97, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25780041

RESUMEN

How the formation and activity of CD4(+)Foxp3(+) regulatory T cells (Tregs) are shaped by TCR recognition of the diverse array of peptide:MHC complexes that can be generated from self-antigens and/or foreign Ags in vivo remains poorly understood. We show that a self-peptide with low (but not high) stimulatory potency promotes thymic Treg formation and can induce conventional CD4(+) T cells in the periphery to become Tregs that express different levels of the transcription factor Helios according to anatomical location. When Tregs generated in response to this self-peptide subsequently encountered the same peptide derived instead from influenza virus in the lung-draining lymph nodes of infected mice, they proliferated, acquired a T-bet(+)CXCR3(+) phenotype, and suppressed the antiviral effector T cell response in the lungs. However, these self-antigen-selected Tregs were unable to suppress the antiviral immune response based on recognition of the peptide as a self-antigen rather than a viral Ag. Notably, when expressed in a more immunostimulatory form, the self-peptide inhibited the formation of T-bet(+)CXCR3(+) Tregs in response to viral Ag, and Ag-expressing B cells from these mice induced Treg division without upregulation of CXCR3. These studies show that a weakly immunostimulatory self-peptide can induce thymic and peripheral Foxp3(+) Treg formation but is unable to activate self-antigen-selected Tregs to modulate an antiviral immune response. Moreover, a strongly immunostimulatory self-peptide expressed by B cells induced Tregs to proliferate without acquiring an effector phenotype that allows trafficking from the draining lymph node to the lungs and, thereby, prevented the Tregs from suppressing the antiviral immune response.


Asunto(s)
Presentación de Antígeno , Diferenciación Celular/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos Virales/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Diferenciación Celular/genética , División Celular/inmunología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/patología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas de Dominio T Box/inmunología , Linfocitos T Reguladores/patología
8.
J Virol ; 88(20): 11995-2005, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100838

RESUMEN

Pathogen-specific antibodies (Abs) protect against respiratory infection with influenza A virus (IAV) and Streptococcus pneumoniae and are the basis of effective vaccines. Sequential or overlapping coinfections with both pathogens are common, yet the impact of coinfection on the generation and maintenance of Ab responses is largely unknown. We report here that the B cell response to IAV is altered in mice coinfected with IAV and S. pneumoniae and that this response differs, depending on the order of pathogen exposure. In mice exposed to S. pneumoniae prior to IAV, the initial virus-specific germinal center (GC) B cell response is significantly enhanced in the lung-draining mediastinal lymph node and spleen, and there is an increase in CD4(+) T follicular helper (TFH) cell numbers. In contrast, secondary S. pneumoniae infection exaggerates early antiviral antibody-secreting cell formation, and at later times, levels of GCs, TFH cells, and antiviral serum IgG are elevated. Mice exposed to S. pneumoniae prior to IAV do not maintain the initially robust GC response in secondary lymphoid organs and exhibit reduced antiviral serum IgG with diminished virus neutralization activity a month after infection. Our data suggest that the history of pathogen exposures can critically affect the generation of protective antiviral Abs and may partially explain the differential susceptibility to and disease outcomes from IAV infection in humans. Importance: Respiratory tract coinfections, specifically those involving influenza A viruses and Streptococcus pneumoniae, remain a top global health burden. We sought to determine how S. pneumoniae coinfection modulates the B cell immune response to influenza virus since antibodies are key mediators of protection.


Asunto(s)
Linfocitos B/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Femenino , Citometría de Flujo , Centro Germinal , Humanos , Gripe Humana/complicaciones , Ratones , Ratones Endogámicos BALB C , Infecciones Neumocócicas/complicaciones
9.
Virology ; 462-463: 254-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24999050

RESUMEN

The response to influenza virus (IAV) infection and severity of disease is highly variable in humans. We hypothesized that one factor contributing to this variability is the presence of specific respiratory tract (RT) microbes. One such microbe is Streptococcus pneumoniae (Sp) that is carried asymptomatically in the RT of many humans. In a mouse co-infection model we found that in contrast to secondary bacterial infection that exacerbates disease, Sp colonization 10 days prior to IAV protects from virus-induced morbidity and lung pathology. Using mutant Sp strains, we identified a critical role for the bacterial virulence factor pneumolysin (PLY) in mediating this protection. Colonization with the PLY-sufficient Sp strain induces expression of the immune-suppressive enzyme arginase 1 in alveolar macrophages (aMø) and correlates with attenuated recruitment and function of pulmonary inflammatory cells. Our study demonstrates a novel role for PLY in Sp-mediated protection by maintaining aMø as "gatekeepers" against virus-induced immunopathology.


Asunto(s)
Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Streptococcus pneumoniae/enzimología , Estreptolisinas/inmunología , Estreptolisinas/metabolismo , Animales , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Análisis de Supervivencia
10.
Nat Immunol ; 15(7): 667-75, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24859450

RESUMEN

CD4(+) follicular helper T cells (T(FH) cells) are essential for germinal center (GC) responses and long-lived antibody responses. Here we report that naive CD4(+) T cells deficient in the transcription factor Foxp1 'preferentially' differentiated into T(FH) cells, which resulted in substantially enhanced GC and antibody responses. We found that Foxp1 used both constitutive Foxp1A and Foxp1D induced by stimulation of the T cell antigen receptor (TCR) to inhibit the generation of T(FH) cells. Mechanistically, Foxp1 directly and negatively regulated interleukin 21 (IL-21); Foxp1 also dampened expression of the costimulatory molecule ICOS and its downstream signaling at early stages of T cell activation, which rendered Foxp1-deficient CD4(+) T cells partially resistant to blockade of the ICOS ligand (ICOSL) during T(FH) cell development. Our findings demonstrate that Foxp1 is a critical negative regulator of T(FH) cell differentiation.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/fisiología , Proteínas Represoras/fisiología , Linfocitos T Colaboradores-Inductores/citología , Animales , Linfocitos T CD4-Positivos/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucinas/genética , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/fisiología
11.
Cell ; 156(4): 705-16, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24529375

RESUMEN

The Toll-like receptors (TLRs) of the innate immune system are unusual in that individual family members are located on different organelles, yet most activate a common signaling pathway important for host defense. It remains unclear how this common signaling pathway can be activated from multiple subcellular locations. Here, we report that, in response to natural activators of innate immunity, the sorting adaptor TIRAP regulates TLR signaling from the plasma membrane and endosomes. TLR signaling from both locations triggers the TIRAP-dependent assembly of the myddosome, a protein complex that controls proinflammatory cytokine expression. The actions of TIRAP depend on the promiscuity of its phosphoinositide-binding domain. Different lipid targets of this domain direct TIRAP to different organelles, allowing it to survey multiple compartments for the presence of activated TLRs. These data establish how promiscuity, rather than specificity, can be a beneficial means of diversifying the subcellular sites of innate immune signal transduction.


Asunto(s)
Inmunidad Innata , Glicoproteínas de Membrana/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Animales , Membrana Celular/metabolismo , Endosomas/metabolismo , Herpes Simple/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores Toll-Like/inmunología
12.
PLoS Pathog ; 9(3): e1003207, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23516357

RESUMEN

Seasonal epidemics of influenza virus result in ∼36,000 deaths annually in the United States. Current vaccines against influenza virus elicit an antibody response specific for the envelope glycoproteins. However, high mutation rates result in the emergence of new viral serotypes, which elude neutralization by preexisting antibodies. T lymphocytes have been reported to be capable of mediating heterosubtypic protection through recognition of internal, more conserved, influenza virus proteins. Here, we demonstrate using a recombinant influenza virus expressing the LCMV GP33-41 epitope that influenza virus-specific CD8+ T cells and virus-specific non-neutralizing antibodies each are relatively ineffective at conferring heterosubtypic protective immunity alone. However, when combined virus-specific CD8 T cells and non-neutralizing antibodies cooperatively elicit robust protective immunity. This synergistic improvement in protective immunity is dependent, at least in part, on alveolar macrophages and/or other lung phagocytes. Overall, our studies suggest that an influenza vaccine capable of eliciting both CD8+ T cells and antibodies specific for highly conserved influenza proteins may be able to provide heterosubtypic protection in humans, and act as the basis for a potential "universal" vaccine.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Macrófagos Alveolares/inmunología , Inmunidad Adaptativa , Animales , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Línea Celular , Protección Cruzada , Perros , Femenino , Glicoproteínas/inmunología , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/inmunología , Carga Viral , Proteínas Virales/inmunología
13.
PLoS One ; 6(12): e28445, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22180783

RESUMEN

BACKGROUND: The extracellular domain of the influenza A virus protein matrix protein 2 (M2e) is remarkably conserved between various human isolates and thus is a viable target antigen for a universal influenza vaccine. With the goal of inducing protection in multiple mouse haplotypes, M2e-based multiple antigenic peptides (M2e-MAP) were synthesized to contain promiscuous T helper determinants from the Plasmodium falciparum circumsporozoite protein, the hepatitis B virus antigen and the influenza virus hemagglutinin. Here, we investigated the nature of the M2e-MAP-induced B cell response in terms of the distribution of antibody (Ab) secreting cells (ASCs) and Ab isotypes, and tested the protective efficacy in various mouse strains. METHODOLOGY/PRINCIPAL FINDINGS: Immunization of BALB/c mice with M2e-MAPs together with potent adjuvants, CpG 1826 oligonucleotides (ODN) and cholera toxin (CT) elicited high M2e-specific serum Ab titers that protected mice against viral challenge. Subcutaneous (s.c.) and intranasal (i.n.) delivery of M2e-MAPs resulted in the induction of IgG in serum and airway secretions, however only i.n. immunization induced anti-M2e IgA ASCs locally in the lungs, correlating with M2-specific IgA in the bronchio-alveolar lavage (BAL). Interestingly, both routes of vaccination resulted in equal protection against viral challenge. Moreover, M2e-MAPs induced cross-reactive and protective responses to diverse M2e peptides and variant influenza viruses. However, in contrast to BALB/c mice, immunization of other inbred and outbred mouse strains did not induce protective Abs. This correlated with a defect in T cell but not B cell responsiveness to the M2e-MAPs. CONCLUSION/SIGNIFICANCE: Anti-M2e Abs induced by M2e-MAPs are highly cross-reactive and can mediate protection to variant viruses. Although synthetic MAPs are promising designs for vaccines, future constructs will need to be optimized for use in the genetically heterogeneous human population.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos B/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Fragmentos de Péptidos/inmunología , Vacunación/métodos , Proteínas de la Matriz Viral/inmunología , Secuencia de Aminoácidos , Animales , Animales no Consanguíneos , Especificidad de Anticuerpos , Antígenos Virales/química , Reacciones Cruzadas , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Mutación , Fragmentos de Péptidos/química , Plasmodium falciparum/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética
14.
J Clin Invest ; 121(10): 3954-64, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21881204

RESUMEN

Antiviral Abs, for example those produced in response to influenza virus infection, are critical for virus neutralization and defense against secondary infection. While the half-life of Abs is short, Ab titers can last a lifetime due to a subset of the Ab-secreting cells (ASCs) that is long lived. However, the mechanisms governing ASC longevity are poorly understood. Here, we have identified a critical role for extrinsic cytokine signals in the survival of respiratory tract ASCs in a mouse model of influenza infection. Irradiation of mice at various time points after influenza virus infection markedly diminished numbers of lung ASCs, suggesting that they are short-lived and require extrinsic factors in order to persist. Neutralization of the TNF superfamily cytokines B lymphocyte stimulator (BLyS; also known as BAFF) and a proliferation-inducing ligand (APRIL) reduced numbers of antiviral ASCs in the lungs and bone marrow, whereas ASCs in the spleen and lung-draining lymph node were surprisingly unaffected. Mice deficient in transmembrane activator and calcium-modulator and cyclophilin ligand interactor (TACI), a receptor for BLyS and APRIL, mounted an initial antiviral B cell response similar to that generated in WT mice but failed to sustain protective Ab titers in the airways and serum, leading to increased susceptibility to secondary viral challenge. These studies highlight the importance of TACI signaling for the maintenance of ASCs and protection against influenza virus infection.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Infecciones por Orthomyxoviridae/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología , Animales , Células Productoras de Anticuerpos/inmunología , Células Productoras de Anticuerpos/patología , Células Productoras de Anticuerpos/efectos de la radiación , Factor Activador de Células B/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Modelos Animales de Enfermedad , Femenino , Pulmón/inmunología , Pulmón/patología , Pulmón/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/deficiencia , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología
15.
J Immunol ; 182(2): 871-9, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124730

RESUMEN

Plasmacytoid dendritic cells (pDC) are thought to be pivotal in the first line of defense against viral infections. Although previous studies have suggested that pDC regulate the immune response against respiratory syncytial virus, their role in pulmonary infection with influenza virus has remained unclear. Using mice with GFP-tagged pDC, we observed a marked increase in pDC numbers in the lung airways 3 days after intranasal infection with influenza virus A/PR/8/34. To further investigate their potential involvement in the disease, we made use of pDC-deficient IkarosL/L mice. In the absence of pDC, the recruitment of T cells to the bronchoalveolar space was delayed, which could be reversed by the adoptive transfer of pDC before infection. Surprisingly, however, when compared with wild-type animals, IkarosL/L mice revealed a similar course of disease, as determined by weight loss, viral titers, levels of neutralizing Ab, and lung pathology. Moreover, the activation and differentiation of influenza-specific CD8+ effector T cells was unaltered in the absence of pDC, as was the generation of CD8+ memory T cells. Taken together, our study suggests that pDC regulate the accumulation of T cells in the bronchoalveolar space during early influenza virus infection, but are dispensable for the control of this disease.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones del Sistema Respiratorio/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/virología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular Tumoral , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Factor de Transcripción Ikaros/genética , Leucocitos/inmunología , Leucocitos/patología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Infecciones por Orthomyxoviridae/patología , Infecciones del Sistema Respiratorio/patología , Linfocitos T/inmunología , Linfocitos T/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...