Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38260519

RESUMEN

During thymic development, most γδ T cells acquire innate-like characteristics that are critical for their function in tumor surveillance, infectious disease, and tissue repair. The mechanisms, however, that regulate γδ T cell developmental programming remain unclear. Recently, we demonstrated that the SLAM-SAP signaling pathway regulates the development and function of multiple innate-like γδ T cell subsets. Here, we used a single-cell proteogenomics approach to identify SAP-dependent developmental checkpoints and to define the SAP-dependent γδ TCR repertoire. SAP deficiency resulted in both a significant loss of an immature Gzma + Blk + Etv5 + Tox2 + γδT17 precursor population, and a significant increase in Cd4 + Cd8 + Rorc + Ptcra + Rag1 + thymic γδ T cells. SAP-dependent diversion of embryonic day 17 thymic γδ T cell clonotypes into the αß T cell developmental pathway was associated with a decreased frequency of mature clonotypes in neonatal thymus, and an altered γδ TCR repertoire in the periphery. Finally, we identify TRGV4/TRAV13-4(DV7)-expressing T cells as a novel, SAP-dependent Vγ4 γδT1 subset. Together, the data suggest that SAP-dependent γδ/αß T cell lineage commitment regulates γδ T cell developmental programming and shapes the γδ TCR repertoire.

2.
J Immunol ; 211(4): 648-657, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37405700

RESUMEN

Drugs are needed to protect against the neutrophil-derived histones responsible for endothelial injury in acute inflammatory conditions such as trauma and sepsis. Heparin and other polyanions can neutralize histones but challenges with dosing or side effects such as bleeding limit clinical application. In this study, we demonstrate that suramin, a widely available polyanionic drug, completely neutralizes the toxic effects of individual histones, but not citrullinated histones from neutrophil extracellular traps. The sulfate groups on suramin form stable electrostatic interactions with hydrogen bonds in the histone octamer with a dissociation constant of 250 nM. In cultured endothelial cells (Ea.Hy926), histone-induced thrombin generation was significantly decreased by suramin. In isolated murine blood vessels, suramin abolished aberrant endothelial cell calcium signals and rescued impaired endothelial-dependent vasodilation caused by histones. Suramin significantly decreased pulmonary endothelial cell ICAM-1 expression and neutrophil recruitment caused by infusion of sublethal doses of histones in vivo. Suramin also prevented histone-induced lung endothelial cell cytotoxicity in vitro and lung edema, intra-alveolar hemorrhage, and mortality in mice receiving a lethal dose of histones. Protection of vascular endothelial function from histone-induced damage is a novel mechanism of action for suramin with therapeutic implications for conditions characterized by elevated histone levels.


Asunto(s)
Histonas , Suramina , Ratones , Animales , Histonas/metabolismo , Suramina/farmacología , Células Endoteliales/metabolismo , Endotelio/metabolismo , Hemorragia
3.
Biomaterials ; 276: 121029, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34311317

RESUMEN

Decellularized pig lungs recellularized with human lung cells offer a novel approach for organ transplantation. However, the potential immunogenicity of decellularized pig lungs following exposure to human tissues has not been assessed. We found that exposure of native lungs from wildtype and transgenic pigs lacking alpha (1,3)-galactosyltransferase (α-gal KO) to sera from normal healthy human volunteers demonstrated similar robust IgM and IgG immunoreactivity, comparably decreased in decellularized lungs. Similar results were observed with sera from patients who had previously undergone transcutaneous porcine aortic valve replacement (TAVR) or from patients with increased circulating anti-α-gal IgE antibodies (α-gal syndrome). Depleting anti-α-gal antibodies from the sera demonstrated both specificity of α-gal immunoreactivity and also residual immunoreactivity similar between wildtype and α-gal KO pig lungs. Exposure of human monocytes and macrophages to native wildtype lungs demonstrated greater induction of M2 phenotype than native α-gal KO pig lungs, which was less marked with decellularized lungs of either type. Overall, these results demonstrate that native wildtype and α-gal KO pig lungs provoke similar immune responses that are comparably decreased following decellularization. This provides a further platform for potential use of decellularized pig lungs in tissue engineering approaches and subsequent transplantation schemes but no obvious overall immunologic advantage of utilizing lungs obtained from α-gal KO pigs.


Asunto(s)
Bioprótesis , Prótesis Valvulares Cardíacas , Animales , Hipersensibilidad a los Alimentos , Galactosiltransferasas/genética , Humanos , Pulmón , Porcinos , Trasplante Heterólogo
4.
Cell Rep Med ; 1(9): 100155, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33377126

RESUMEN

The tetravalent live attenuated dengue vaccine candidate TV003 induces neutralizing antibodies against all four dengue virus serotypes (DENV1-DENV4) and protects against experimental challenge with DENV2 in humans. Here, we track vaccine viremia and B and T cell responses to this vaccination/challenge model to understand how vaccine viremia links adaptive immunity and development of protective antibody responses. TV003 viremia triggers an acute plasmablast response that, in combination with DENV-specific CD4+ T cells, correlates with serum neutralizing antibodies. TV003 vaccinees develop DENV2-reactive memory B cells, including serotype-specific and multivalent specificities in line with the composition of serum antibodies. There is no post-challenge plasmablast response in vaccinees, although stronger and earlier post-TV003 plasmablast responses associate with sterile humoral protection from DENV2 challenge. TV003 vaccine triggers plasmablasts and memory B cells, which, with support from CD4+ T cells, functionally link early vaccine viremia and the serum antibody responses.


Asunto(s)
Linfocitos B/inmunología , Vacunas contra el Dengue/inmunología , Flavivirus/inmunología , Vacunas Atenuadas/inmunología , Inmunidad Adaptativa/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Dengue/inmunología , Virus del Dengue/inmunología , Humanos , Células Plasmáticas/inmunología
5.
Front Immunol ; 11: 479, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32265929

RESUMEN

Dengue virus (DENV) is a mosquito-borne flavivirus that causes serious human disease. The current lack of an effective vaccine to simultaneously protect against the four serotypes of DENV in seronegative individuals is a major unmet medical need. Further, the immunological basis for protective immunity in the setting of DENV infection or vaccination is not fully understood. Our team has developed a live attenuated tetravalent dengue virus vaccine that provides complete protection in a human model of dengue virus challenge. The goal of this study was to define, in the context of protective human vaccination, the quality of vaccine-induced DENV-specific CD8+ and CD4+ T cells and the temporal dynamics associated with their formation and maintenance. Multifunctional, DENV-specific CD8+ and CD4+ T cells developed 8-14 days after vaccination and were maintained for at least 6 months. Virus-specific CD8 T+ cells were a mixture of effector memory T cells (TEM) and effector memory T cells re-expressing CD45RA (TEMRA), with TEM cells predominating until day 21 post-vaccination and TEMRA cells thereafter. The majority of virus-specific CD4+ T cells were TEM with a small fraction being TEMRA. The frequency of virus-specific CD8+ and CD4+ T cells were further skewed to the TEMRA phenotype following either a second dose of the tetravalent vaccine or challenge with a single serotype of DENV. Collectively, our study has defined the phenotypic profile of antiviral CD8+ and CD4+ T cells associated with protective immunity to DENV infection and the kinetics of their formation and maintenance.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Dengue/inmunología , Virus del Dengue/inmunología , Antígenos Comunes de Leucocito/análisis , Subgrupos Linfocitarios/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/química , Linfocitos T CD8-positivos/química , Ensayos Clínicos Fase I como Asunto , Citocinas/análisis , Virus del Dengue/genética , Epítopos/inmunología , Humanos , Inmunización Secundaria , Inmunogenicidad Vacunal , Memoria Inmunológica , Subgrupos Linfocitarios/química , Factores de Tiempo , Vacunación , Vacunas Atenuadas/inmunología
6.
J Immunol ; 204(6): 1521-1534, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32024701

RESUMEN

During thymic development, mouse γδ T cells commit to either an IFN-γ- or an IL-17-producing phenotype through mechanisms that remain unclear. In this study, we investigated the extent to which the SLAM/SAP signaling pathway regulates the functional programming of γδ T cells. Characterization of SLAM family receptor expression revealed that thymic γδ T cell subsets were each marked by distinct coexpression profiles of SLAMF1, SLAMF4, and SLAMF6. In the thymus, Vγ1 and Vγ4 T cells that exhibited an SLAMF1+SLAMF6+ double positive phenotype were largely contained within immature CD24+CD73- and CD24+CD73+ subsets, whereas SLAMF1 single positive, SLAMF6 single positive, or SLAMF1SLAMF6 double negative cells were found within mature CD24-CD73+ and CD24-CD73- subsets. In the periphery, SLAMF1 and SLAMF6 expression distinguished IL-17- and IFN-γ-producing γδ T cells, respectively. Disruption of SLAM family receptor signaling through deletion of SAP resulted in impaired thymic Vγ1 and Vγ4 T cell maturation at the CD24+CD73-SLAMF1+SLAMF6+ double positive stage that was associated with a decreased frequency of CD44+RORγt+ γδ T cells. Impaired development was in turn associated with decreased γδ T cell IL-17 and IFN-γ production in the thymus as well as in peripheral tissues. The role for SAP was subset-specific, as Vγ1Vδ6.3, Vγ4, Vγ5, but not Vγ6 subsets were SAP-dependent. Together, these data suggest that the SLAM/SAP signaling pathway plays a larger role in γδ T cell development than previously appreciated and represents a critical checkpoint in the functional programming of both IL-17- and IFN-γ-producing γδ T cell subsets.


Asunto(s)
Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Subgrupos de Linfocitos T/metabolismo , Timo/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Masculino , Ratones , Modelos Animales , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/genética , Subgrupos de Linfocitos T/inmunología , Timo/citología , Timo/inmunología
7.
Genes Immun ; 20(4): 261-272, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29880961

RESUMEN

Invariant NKT (iNKT) cells are tissue-resident innate-like T cells critical to the host immune response. We previously identified a 6.6 Mbp region on chromosome 1 as a major regulator of iNKT cell number and function in C57BL/6 and 129X1/SvJ mice. Here, we fine-mapped this locus by assessing the iNKT cell response to alpha-galactosylceramide (αGalCer) in a series of B6.129 congenic lines. This analysis revealed the presence of at least two genetic elements that regulate iNKT cell cytokine production in response to αGalCer. While one of these genetic elements mapped to the B6.129c6 interval containing Slam genes, the dominant regulator in this region mapped to the 0.14 Mbp B6.129c3 interval. In addition, we found that numbers of thymic iNKT cells and DP thymocytes were significantly lower in B6.129c3 mice, indicating that this interval also regulates iNKT cell development. Candidate gene analysis revealed a fivefold increase in Fcgr3 expression in B6.129c3 iNKT cells, and we observed increased expression of FcγR3 protein on B6.129c3 iNKT cells, NK cells, and neutrophils. These data identify the B6.129c3 interval as a novel locus regulating the response of iNKT cells to glycosphingolipid, revealing a link between this phenotype and a polymorphism that regulates Fcgr3 expression.


Asunto(s)
Sitios Genéticos , Inmunidad Innata/genética , Células Asesinas Naturales/inmunología , Receptores de IgG/genética , Animales , Células Cultivadas , Citocinas/metabolismo , Galactosilceramidas/farmacología , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Receptores de IgG/metabolismo
8.
Sci Rep ; 8(1): 16571, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30410021

RESUMEN

Serum amyloid A (SAA) proteins are a family of acute phase apolipoproteins implicated to directly modulate innate and adaptive immune responses. However, new studies comparing endogenous SAAs and recombinant forms of these proteins have questioned the function of SAA in inflammation and immunity. We generated SAA3 knockout mice to evaluate the contribution of SAA3 to lung development and immune-mediated lung disease. While SAA3 deficiency does not affect the generation of house dust mite-induced allergic asthma, mice lacking SAA3 develop adult-onset obesity, intrinsic airway hyperresponsiveness, increased inflammatory and fibrotic gene expression in the lung, and elevated levels of lung citrullinated proteins. Polyclonally stimulated CD4+ T cells from SAA3-/- mice exhibit impaired glycolytic activity, decreased TH2 and TH1 cytokine secretion, and elevated IL-17A production compared to wild type cells. Polyclonally stimulated CD8+ T cells from SAA3-/- mice also exhibit impaired glycolytic activity as well as a diminished capacity to produce IL-2 and IFNγ. Finally, SAA3-/- mice demonstrate increased mortality in response to H1N1 influenza infection, along with higher copy number of viral RNAs in the lung, a lack of CD8+ T cell IFNγ secretion, and decreased flu-specific antibodies. Our findings indicate that endogenous SAA3 regulates lung development and homeostasis, and is required for protection against H1N1 influenza infection.


Asunto(s)
Enfermedades Pulmonares/virología , Pulmón/crecimiento & desarrollo , Infecciones por Orthomyxoviridae/genética , Hipersensibilidad Respiratoria/genética , Proteína Amiloide A Sérica/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Técnicas de Inactivación de Genes , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Lipopolisacáridos/efectos adversos , Pulmón/metabolismo , Pulmón/virología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/patología , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Pyroglyphidae/inmunología , Hipersensibilidad Respiratoria/inmunología , Proteína Amiloide A Sérica/metabolismo , Análisis de Supervivencia
9.
J Immunol ; 201(8): 2377-2384, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30158125

RESUMEN

Studies comparing endogenous and recombinant serum amyloid A (SAA) have generated conflicting data on the proinflammatory function of these proteins. In exploring this discrepancy, we found that in contrast to commercially sourced recombinant human SAA1 (hSAA1) proteins produced in Escherichia coli, hSAA1 produced from eukaryotic cells did not promote proinflammatory cytokine production from human or mouse cells, induce Th17 differentiation, or stimulate TLR2. Proteomic analysis of E. coli-derived hSAA1 revealed the presence of numerous bacterial proteins, with several being reported or probable lipoproteins. Treatment of hSAA1 with lipoprotein lipase or addition of a lipopeptide to eukaryotic cell-derived hSAA1 inhibited or induced the production of TNF-α from macrophages, respectively. Our results suggest that a function of SAA is in the binding of TLR2-stimulating bacterial proteins, including lipoproteins, and demand that future studies of SAA employ a recombinant protein derived from eukaryotic cells.


Asunto(s)
Leucocitos Mononucleares/inmunología , Proteína Amiloide A Sérica/inmunología , Células Th17/inmunología , Receptor Toll-Like 2/agonistas , Adulto , Animales , Diferenciación Celular , Citocinas/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/inmunología , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Lipoproteínas/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteína Amiloide A Sérica/genética
10.
Cell Death Dis ; 9(2): 62, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29352186

RESUMEN

Resting T cells undergo a rapid metabolic shift to glycolysis upon activation in the presence of interleukin (IL)-2, in contrast to oxidative mitochondrial respiration with IL-15. Paralleling these different metabolic states are striking differences in susceptibility to restimulation-induced cell death (RICD); glycolytic effector T cells are highly sensitive to RICD, whereas non-glycolytic T cells are resistant. It is unclear whether the metabolic state of a T cell is linked to its susceptibility to RICD. Our findings reveal that IL-2-driven glycolysis promotes caspase-3 activity and increases sensitivity to RICD. Neither caspase-7, caspase-8, nor caspase-9 activity is affected by these metabolic differences. Inhibition of glycolysis with 2-deoxyglucose reduces caspase-3 activity as well as sensitivity to RICD. By contrast, IL-15-driven oxidative phosphorylation actively inhibits caspase-3 activity through its glutathionylation. We further observe active caspase-3 in the lipid rafts of glycolytic but not non-glycolytic T cells, suggesting a proximity-induced model of self-activation. Finally, we observe that effector T cells during influenza infection manifest higher levels of active caspase-3 than naive T cells. Collectively, our findings demonstrate that glycolysis drives caspase-3 activity and susceptibility to cell death in effector T cells independently of upstream caspases. Linking metabolism, caspase-3 activity, and cell death provides an intrinsic mechanism for T cells to limit the duration of effector function.


Asunto(s)
Caspasa 3/metabolismo , Glucólisis/genética , Linfocitos T/metabolismo , Humanos
11.
Pulm Pharmacol Ther ; 45: 80-89, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28483562

RESUMEN

iNKT cells and mast cells have both been implicated in the syndrome of allergic asthma through their activation-induced release of Th2 type cytokines and secretion of histamine and other mediators, respectively, which can promote airways hyperresponsiveness (AHR) to agents such as methacholine. However, a mechanistic link between iNKT cells and mast cell recruitment or activation has never been explored. Our objective was to determine whether iNKT cells are necessary for the recruitment of mast cells and if iNKT cells can influence the acute allergen induced bronchoconstriction (AIB) caused by mast cell mediator release. To do so, we pharmacologically eliminated iNKT cells using a specific antibody (NKT-14) and examined its impact on airway inflammation and physiological phenotype. In mice treated with NKT-14, the elimination of iNKT cells was sufficient to prevent AHR and pulmonary eosinophilic inflammation elicited by administration of the iNKT cell agonist αGalCer. In mice treated with NKT-14 and then sensitized and challenged with house dust mite extract (HDM), eliminating the iNKT cells significantly reduced both AHR and AIB but did not affect pulmonary inflammation, the mast cell population, nor the release of the mast cell mediators mast cell protease-1 and prostaglandin D2. We conclude that while iNKT cells contribute to the phenotype of allergic airways disease through the manifestation of AIB and AHR, their presence is not required for mast cell recruitment and activation, or to generate the characteristic inflammatory response subsequent to allergen challenge.


Asunto(s)
Broncoconstricción/inmunología , Mastocitos/metabolismo , Células T Asesinas Naturales/metabolismo , Hipersensibilidad Respiratoria/inmunología , Alérgenos/inmunología , Animales , Quimasas/metabolismo , Modelos Animales de Enfermedad , Eosinófilos/metabolismo , Femenino , Hipersensibilidad/inmunología , Inflamación/inmunología , Pulmón/inmunología , Pulmón/patología , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Células T Asesinas Naturales/inmunología , Fenotipo , Prostaglandina D2/metabolismo , Pyroglyphidae/inmunología
12.
Proc Natl Acad Sci U S A ; 114(13): 3491-3496, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28242695

RESUMEN

Males of many species, ranging from humans to insects, are more susceptible than females to parasitic, fungal, bacterial, and viral infections. One mechanism that has been proposed to account for this difference is the immunocompetence handicap model, which posits that the greater infectious disease burden in males is due to testosterone, which drives the development of secondary male sex characteristics at the expense of suppressing immunity. However, emerging data suggest that cell-intrinsic (chromosome X and Y) sex-specific factors also may contribute to the sex differences in infectious disease burden. Using a murine model of influenza A virus (IAV) infection and a panel of chromosome Y (ChrY) consomic strains on the C57BL/6J background, we present data showing that genetic variation in ChrY influences IAV pathogenesis in males. Specific ChrY variants increase susceptibility to IAV in males and augment pathogenic immune responses in the lung, including activation of proinflammatory IL-17-producing γδ T cells, without affecting viral replication. In addition, susceptibility to IAV segregates independent of copy number variation in multicopy ChrY gene families that influence susceptibility to other immunopathological phenotypes, including survival after infection with coxsackievirus B3. These results demonstrate a critical role for genetic variation in ChrY in regulating susceptibility to infectious disease.


Asunto(s)
Virus de la Influenza A/fisiología , Gripe Humana/genética , Cromosoma Y/genética , Animales , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Gripe Humana/inmunología , Gripe Humana/virología , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Factores Sexuales , Linfocitos T/inmunología , Virulencia
13.
J Immunol ; 197(5): 1720-32, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27465529

RESUMEN

Nitrogen dioxide (NO2) is an environmental air pollutant and endogenously generated oxidant that contributes to the exacerbation of respiratory disease and can function as an adjuvant to allergically sensitize to an innocuous inhaled Ag. Because uric acid has been implicated as a mediator of adjuvant activity, we sought to determine whether uric acid was elevated and participated in a mouse model of NO2-promoted allergic sensitization. We found that uric acid was increased in the airways of mice exposed to NO2 and that administration of uricase inhibited the development of OVA-driven allergic airway disease subsequent to OVA challenge, as well as the generation of OVA-specific Abs. However, uricase was itself immunogenic, inducing a uricase-specific adaptive immune response that occurred even when the enzymatic activity of uricase had been inactivated. Inhibition of the OVA-specific response was not due to the capacity of uricase to inhibit the early steps of OVA uptake or processing and presentation by dendritic cells, but occurred at a later step that blocked OVA-specific CD4(+) T cell proliferation and cytokine production. Although blocking uric acid formation by allopurinol did not affect outcomes, administration of ultra-clean human serum albumin at protein concentrations equivalent to that of uricase inhibited NO2-promoted allergic airway disease. These results indicate that, although uric acid levels are elevated in the airways of NO2-exposed mice, the powerful inhibitory effect of uricase administration on allergic sensitization is mediated more through Ag-specific immune deviation than via suppression of allergic sensitization, a mechanism to be considered in the interpretation of results from other experimental systems.


Asunto(s)
Asma/prevención & control , Hipersensibilidad/inmunología , Dióxido de Nitrógeno/toxicidad , Ovalbúmina/inmunología , Urato Oxidasa/administración & dosificación , Ácido Úrico/metabolismo , Inmunidad Adaptativa , Alérgenos/administración & dosificación , Alopurinol/administración & dosificación , Animales , Presentación de Antígeno , Asma/inducido químicamente , Asma/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Pulmón/química , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/administración & dosificación , Albúmina Sérica/administración & dosificación , Células Th2 , Urato Oxidasa/metabolismo
14.
PLoS One ; 10(10): e0140729, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26474487

RESUMEN

Invariant Natural Killer T (iNKT) cells are a T cell subset expressing an invariant T Cell Receptor (TCR) that recognizes glycolipid antigens rather than peptides. The cells have both innate-like rapid cytokine release, and adaptive-like thymic positive selection. iNKT cell activation has been implicated in the pathogenesis of allergic asthma and inflammatory diseases, while reduced iNKT cell activation promotes infectious disease, cancer and certain autoimmune diseases such as Type 1 diabetes (T1D). Therapeutic means to reduce or deplete iNKT cells could treat inflammatory diseases, while approaches to promote their activation may have potential in certain infectious diseases, cancer or autoimmunity. Thus, we developed invariant TCR-specific monoclonal antibodies to better understand the role of iNKT cells in disease. We report here the first monoclonal antibodies specific for the mouse invariant TCR that by modifying the Fc construct can specifically deplete or activate iNKT cells in vivo in otherwise fully immuno-competent animals. We have used both the depleting and activating version of the antibody in the NOD model of T1D. As demonstrated previously using genetically iNKT cell deficient NOD mice, and in studies of glycolipid antigen activated iNKT cells in standard NOD mice, we found that antibody mediated depletion or activation of iNKT cells respectively accelerated and retarded T1D onset. In BALB/c mice, ovalbumin (OVA) mediated airway hyper-reactivity (AHR) was abrogated with iNKT cell depletion prior to OVA sensitization, confirming studies in knockout mice. Depletion of iNKT cells after sensitization had no effect on AHR in the conducting airways but did reduce AHR in the lung periphery. This result raises caution in the interpretation of studies that use animals that are genetically iNKT cell deficient from birth. These activating and depleting antibodies provide a novel tool to assess the therapeutic potential of iNKT cell manipulation.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Asma/inmunología , Diabetes Mellitus Tipo 1/inmunología , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales de Origen Murino/inmunología , Asma/genética , Asma/patología , Asma/terapia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Depleción Linfocítica/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Células T Asesinas Naturales/patología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
15.
Infect Immun ; 83(6): 2557-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870224

RESUMEN

Pseudomonas aeruginosa is an important human opportunistic pathogen, accounting for a significant fraction of hospital-acquired lung infections. CD1d-restricted NKT cells comprise an unusual innate-like T cell subset that plays important roles in both bacterial and viral infections. Previous reports have differed in their conclusions regarding the role of NKT cells in clearance of P. aeruginosa from the lung. Since there is significant strain-dependent variation in NKT cell number and function among different inbred strains of mice, we investigated whether the role of NKT cells was dependent on the host genetic background. We found that NKT cells did indeed play a critical role in the clearance of P. aeruginosa from the lungs of BALB/c mice but that they played no discernible role in clearance from the lungs of C57BL/6 mice. We found that the strain-dependent role of NKT cells was associated with significant strain-dependent differences in cytokine production by lung NKT cells and that impaired clearance of P. aeruginosa in BALB/c CD1d(-/-) mice was associated with an increase in neutrophil influx to the lung and increased levels of proinflammatory cytokines and chemokines after infection. Finally, we found that the role of alveolar macrophages was also dependent on the genetic background. These data provide further support for a model in which the unusually high level of variability in NKT cell number and function among different genetic backgrounds may be an important contributor to infectious-disease susceptibility and pathology.


Asunto(s)
Antígenos CD1d/metabolismo , Pulmón/citología , Células T Asesinas Naturales/fisiología , Neumonía Bacteriana/microbiología , Pseudomonas aeruginosa/fisiología , Animales , Antígenos CD1d/genética , Lavado Broncoalveolar , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Humanos , Pulmón/microbiología , Macrófagos Alveolares , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neumonía Bacteriana/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo
16.
J Immunol ; 192(1): 189-99, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24307737

RESUMEN

CD1d-restricted NKT cells comprise an innate-like T cell population that exerts significant influence over early events in the developing immune response. The frequency of NKT cells is highly variable in humans and in mice, but the basis for this variability remains unclear. In this study, we report a striking deficiency of type I NKT cells in the wild-derived inbred strains PWD/PhJ, SPRET/EiJ, and CAST/EiJ. Investigation of the underlying basis for the lack of type I NKT cells revealed that one strain, PWD/PhJ, exhibited a significant impairment in thymocyte and splenocyte CD1d gene and protein expression. Accordingly, both thymocytes and bone marrow-derived dendritic cells from PWD mice exhibited a significant impairment in the ability to present α-galactosylceramide to NKT cells. The impaired PWD CD1d gene expression was due to impaired CD1d promoter activity. Fine-mapping of the promoter activity revealed that two single nucleotide substitutions at positions -331 and -164 in the proximal promoter were each sufficient to account for the diminished PWD CD1d promoter activity. Examination of the strain distribution pattern of these polymorphisms revealed that, of 19 strains analyzed, only PWD and PWK mice possessed both CD1d promoter polymorphisms. A subsequent examination of the PWK strain revealed that it also exhibited impaired thymocyte CD1d expression and very low numbers of NKT cells. Taken together, these results provide new insight into the control of CD1d gene expression, and they have implications for the evolution of CD1d and type I NKT cells.


Asunto(s)
Antígenos CD1d/genética , Regulación de la Expresión Génica , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Polimorfismo Genético , Regiones Promotoras Genéticas , Animales , Presentación de Antígeno/inmunología , Ratones , Células T Asesinas Naturales/inmunología , Polimorfismo de Nucleótido Simple , Timocitos/inmunología , Timocitos/metabolismo
17.
Cell Host Microbe ; 14(5): 522-34, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24237698

RESUMEN

Arenaviruses and hantaviruses cause severe human disease. Little is known regarding host proteins required for their propagation. We identified human proteins that interact with the glycoproteins (GPs) of a prototypic arenavirus and hantavirus and show that the lectin endoplasmic reticulum (ER)-Golgi intermediate compartment 53 kDa protein (ERGIC-53), a cargo receptor required for glycoprotein trafficking within the early exocytic pathway, associates with arenavirus, hantavirus, coronavirus, orthomyxovirus, and filovirus GPs. ERGIC-53 binds to arenavirus GPs through a lectin-independent mechanism, traffics to arenavirus budding sites, and is incorporated into virions. ERGIC-53 is required for arenavirus, coronavirus, and filovirus propagation; in its absence, GP-containing virus particles form but are noninfectious, due in part to their inability to attach to host cells. Thus, we have identified a class of pathogen-derived ERGIC-53 ligands, a lectin-independent basis for their association with ERGIC-53, and a role for ERGIC-53 in the propagation of several highly pathogenic RNA virus families.


Asunto(s)
Arenavirus/fisiología , Coronavirus/fisiología , Filoviridae/fisiología , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/metabolismo , Ensamble de Virus , Línea Celular , Glicoproteínas/metabolismo , Humanos , Transporte de Proteínas , Proteínas Virales/metabolismo
18.
Am J Pathol ; 183(2): 441-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23746656

RESUMEN

Coxsackievirus B3 (CVB3) variants H3 and H310A1 differ by a single nonconserved amino acid in the VP2 capsid region. C57Bl/6 mice infected with the H3 virus develop myocarditis correlating with activation of T cells expressing the Vγ4 T cell receptor chain. Infecting mice with H310A1 activates natural killer T (NKT; mCD1d-tetramer(+) TCRß(+)) cells, but not Vγ4 T cells, and fails to induce myocarditis. H310A1 infection preferentially activates M2 alternatively activated macrophage and CD4(+)FoxP3 (T regulatory) cells, whereas CD4(+)Th1 (IFN-γ(+)) cells are suppressed. By contrast, H3 virus infection activates M1 proinflammatory and CD4(+)Th1 cells, but not T regulatory cells. The M1 macrophage show significantly increased CD1d expression compared to M2 macrophage. The ability of NKT cells to suppress myocarditis was shown by adoptive transfer of purified NKT cells into H3-infected NKT knockout (Jα18 knockout) mice, which inhibited cardiac inflammation and increased T regulatory cell response. Cardiac virus titers were equivalent in all mouse strains indicating that neither Vγ4 nor NKT cells participate in control of virus infection. These data show that NKT and Vγ4 cells cross-regulate T regulatory cell responses during CVB3 infections and are the primary factor determining viral pathogenesis in this mouse model.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Enterovirus Humano B/inmunología , Células T Asesinas Naturales/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD1/metabolismo , Linfocitos T CD4-Positivos/inmunología , Inmunidad Innata/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/virología , Linfocitos T Reguladores/virología
19.
Am J Respir Cell Mol Biol ; 48(5): 655-64, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23371061

RESUMEN

Nitrogen dioxide (NO2) is an environmental pollutant and endogenously generated oxidant associated with the development, severity, and exacerbation of asthma. NO2 exposure is capable of allergically sensitizing mice to the innocuous inhaled antigen ovalbumin (OVA), promoting neutrophil and eosinophil recruitment, and a mixed Th2/Th17 response upon antigen challenge that is reminiscent of severe asthma. However, the identity of IL-17A-producing cells and the mechanisms governing their ontogeny in NO2-promoted allergic airway disease remain unstudied. We measured the kinetics of lung inflammation after antigen challenge in NO2-promoted allergic airway disease, including inflammatory cells in bronchoalveolar lavage and antigen-specific IL-17A production from the lung. We determined that IL-17A(+) cells were predominately CD4(+)T cell receptor (TCR)ß(+) Th17 cells, and that a functional IL-1 receptor was required for Th17, but not Th2, cytokine production after in vitro antigen restimulation of lung cells. The absence of natural killer T cells, γδ T cells, or the inflammasome scaffold nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain (Nlrp)3 did not affect the development of NO2-promoted allergic inflammation or IL-17A production. Similarly, neutrophil depletion or the neutralization of IL-1α during sensitization exerted no effect on these parameters. However, the absence of caspase-1 significantly reduced IL-17A production from lung cells without affecting Th2 cytokines or lung inflammation. Finally, the intranasal administration of IL-1ß and the inhalation of antigen promoted allergic sensitization that was reflected by neutrophilic airway inflammation and IL-17A production from CD4(+)TCRß(+) Th17 cells subsequent to antigen challenge. These data implicate a role for caspase-1 and IL-1ß in the IL-1 receptor-dependent Th17 response manifest in NO2-promoted allergic airway disease.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Asma/metabolismo , Caspasa 1/metabolismo , Dióxido de Nitrógeno/toxicidad , Receptores de Interleucina-1/metabolismo , Células Th17/metabolismo , Animales , Asma/inducido químicamente , Asma/inmunología , Eosinófilos/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Cinética , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Interleucina-1/genética , Bazo/inmunología , Bazo/metabolismo , Células Th17/inmunología
20.
Am J Pathol ; 182(2): 401-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23195432

RESUMEN

There are two major haplotypes of signal lymphocytic activation molecule (Slam) in inbred mouse strains, with the Slam haplotype 1 expressed in C57Bl/6 mice and the Slam haplotype 2 expressed in most other commonly used inbred strains, including 129 mice. Because signaling through Slam family receptors can affect innate immunity [natural killer T cell (NKT) and γ-δ T-cell receptor], and innate immunity can determine susceptibility to coxsackievirus B3 (CVB3) infection, the present study evaluated the response of C57Bl/6 and congenic B6.129c1 mice (expressing the 129-derived Slam locus) to CVB3. CVB3-infected C57Bl/6 male mice developed increased myocarditis but reduced hepatic injury compared with infected B6.129c1 mice. C57Bl/6 mice also had increased γδ(+) and CD8(+)interferon-γ(+) cells but decreased numbers of NKT (T-cell receptor ß chain + mCD1d tetramer(+)) and CD4(+)FoxP3(+) cells compared with B6.129c1 mice. C57Bl/6 mice were infected with CVB3 and treated with either α-galactosylceramide, an NKT cell-specific ligand, or vehicle (dimethyl sulfoxide/PBS). Mice treated with α-galactosylceramide showed significantly reduced myocarditis. Liver injuries, as determined by alanine aminotransferase levels in plasma, were increased significantly, confirming that NKT cells are protective for myocarditis but pathogenic in the liver.


Asunto(s)
Antígenos CD/metabolismo , Infecciones por Coxsackievirus/inmunología , Haplotipos/genética , Hígado/patología , Activación de Linfocitos/inmunología , Miocarditis/patología , Células T Asesinas Naturales/inmunología , Receptores de Superficie Celular/metabolismo , Inmunidad Adaptativa/efectos de los fármacos , Alanina Transaminasa/sangre , Animales , Antígenos CD/genética , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/patología , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/inmunología , Galactosilceramidas/farmacología , Hepatitis/complicaciones , Hepatitis/inmunología , Hepatitis/patología , Hígado/inmunología , Hígado/virología , Activación de Linfocitos/efectos de los fármacos , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocarditis/sangre , Miocarditis/complicaciones , Miocarditis/inmunología , Células T Asesinas Naturales/efectos de los fármacos , Polimorfismo Genético , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Superficie Celular/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Troponina I/sangre , Carga Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...