Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Immunol ; 211(4): 648-657, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37405700

RESUMO

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.


Assuntos
Histonas , Suramina , Camundongos , Animais , Histonas/metabolismo , Suramina/farmacologia , Células Endoteliais/metabolismo , Endotélio/metabolismo , Hemorragia
2.
J Immunol ; 204(6): 1521-1534, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-32024701

RESUMO

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.


Assuntos
Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Subpopulações de Linfócitos T/metabolismo , Timo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Interferon gama/metabolismo , Interleucina-17/metabolismo , Masculino , Camundongos , Modelos Animais , Cultura Primária de Células , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/genética , Subpopulações de Linfócitos T/imunologia , Timo/citologia , Timo/imunologia
3.
J Immunol ; 201(8): 2377-2384, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30158125

RESUMO

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.


Assuntos
Leucócitos Mononucleares/imunologia , Proteína Amiloide A Sérica/imunologia , Células Th17/imunologia , Receptor 2 Toll-Like/agonistas , Adulto , Animais , Diferenciação Celular , Citocinas/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Lipoproteínas/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/genética , Proteína Amiloide A Sérica/genética
4.
Proc Natl Acad Sci U S A ; 114(13): 3491-3496, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28242695

RESUMO

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.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/genética , Cromossomo Y/genética , Animais , Feminino , Predisposição Genética para Doença , Variação Genética , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Influenza Humana/imunologia , Influenza Humana/virologia , Interleucina-17/genética , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Fatores Sexuais , Linfócitos T/imunologia , Virulência
5.
Genes Immun ; 20(4): 261-272, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29880961

RESUMO

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.


Assuntos
Loci Gênicos , Imunidade Inata/genética , Células Matadoras Naturais/imunologia , Receptores de IgG/genética , Animais , Células Cultivadas , Citocinas/metabolismo , Galactosilceramidas/farmacologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Receptores de IgG/metabolismo
6.
J Immunol ; 197(5): 1720-32, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27465529

RESUMO

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.


Assuntos
Asma/prevenção & controle , Hipersensibilidade/imunologia , Dióxido de Nitrogênio/toxicidade , Ovalbumina/imunologia , Urato Oxidase/administração & dosagem , Ácido Úrico/metabolismo , Imunidade Adaptativa , Alérgenos/administração & dosagem , Alopurinol/administração & dosagem , Animais , Apresentação de Antígeno , Asma/induzido quimicamente , Asma/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Humanos , Pulmão/química , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/administração & dosagem , Albumina Sérica/administração & dosagem , Células Th2 , Urato Oxidase/metabolismo
7.
Pulm Pharmacol Ther ; 45: 80-89, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28483562

RESUMO

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.


Assuntos
Broncoconstrição/imunologia , Mastócitos/metabolismo , Células T Matadoras Naturais/metabolismo , Hipersensibilidade Respiratória/imunologia , Alérgenos/imunologia , Animais , Quimases/metabolismo , Modelos Animais de Doenças , Eosinófilos/metabolismo , Feminino , Hipersensibilidade/imunologia , Inflamação/imunologia , Pulmão/imunologia , Pulmão/patologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células T Matadoras Naturais/imunologia , Fenótipo , Prostaglandina D2/metabolismo , Pyroglyphidae/imunologia
8.
J Immunol ; 192(1): 189-99, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24307737

RESUMO

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.


Assuntos
Antígenos CD1d/genética , Regulação da Expressão Gênica , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/metabolismo , Polimorfismo Genético , Regiões Promotoras Genéticas , Animais , Apresentação de Antígeno/imunologia , Camundongos , Células T Matadoras Naturais/imunologia , Polimorfismo de Nucleotídeo Único , Timócitos/imunologia , Timócitos/metabolismo
9.
Infect Immun ; 83(6): 2557-65, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25870224

RESUMO

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.


Assuntos
Antígenos CD1d/metabolismo , Pulmão/citologia , Células T Matadoras Naturais/fisiologia , Pneumonia Bacteriana/microbiologia , Pseudomonas aeruginosa/fisiologia , Animais , Antígenos CD1d/genética , Lavagem Broncoalveolar , Células Cultivadas , Regulação da Expressão Gênica/imunologia , Humanos , Pulmão/microbiologia , Macrófagos Alveolares , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Pneumonia Bacteriana/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo
10.
Am J Pathol ; 182(2): 401-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23195432

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Infecções por Coxsackievirus/imunologia , Haplótipos/genética , Fígado/patologia , Ativação Linfocitária/imunologia , Miocardite/patologia , Células T Matadoras Naturais/imunologia , Receptores de Superfície Celular/metabolismo , Imunidade Adaptativa/efeitos dos fármacos , Alanina Transaminase/sangue , Animais , Antígenos CD/genética , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/patologia , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/imunologia , Galactosilceramidas/farmacologia , Hepatite/complicações , Hepatite/imunologia , Hepatite/patologia , Fígado/imunologia , Fígado/virologia , Ativação Linfocitária/efeitos dos fármacos , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/sangue , Miocardite/complicações , Miocardite/imunologia , Células T Matadoras Naturais/efeitos dos fármacos , Polimorfismo Genético , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Superfície Celular/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Troponina I/sangue , Carga Viral/imunologia
11.
Am J Pathol ; 183(2): 441-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23746656

RESUMO

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.


Assuntos
Infecções por Coxsackievirus/imunologia , Enterovirus Humano B/imunologia , Células T Matadoras Naturais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD1/metabolismo , Linfócitos T CD4-Positivos/imunologia , Imunidade Inata/imunologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/virologia , Linfócitos T Reguladores/virologia
12.
bioRxiv ; 2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38260519

RESUMO

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.

13.
Am J Respir Cell Mol Biol ; 48(5): 655-64, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23371061

RESUMO

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.


Assuntos
Poluentes Atmosféricos/toxicidade , Asma/metabolismo , Caspase 1/metabolismo , Dióxido de Nitrogênio/toxicidade , Receptores de Interleucina-1/metabolismo , Células Th17/metabolismo , Animais , Asma/induzido quimicamente , Asma/imunologia , Eosinófilos/imunologia , Mediadores da Inflamação/metabolismo , Interleucina-17/metabolismo , Cinética , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Interleucina-1/genética , Baço/imunologia , Baço/metabolismo , Células Th17/imunologia
14.
J Immunol ; 187(1): 64-73, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622869

RESUMO

IL-1ß is a cytokine critical to several inflammatory diseases in which pathogenic Th17 responses are implicated. Activation of the NLRP3 inflammasome by microbial and environmental stimuli can enable the caspase-1-dependent processing and secretion of IL-1ß. The acute-phase protein serum amyloid A (SAA) is highly induced during inflammatory responses, wherein it participates in systemic modulation of innate and adaptive immune responses. Elevated levels of IL-1ß, SAA, and IL-17 are present in subjects with severe allergic asthma, yet the mechanistic relationship among these mediators has yet to be identified. In this study, we demonstrate that Saa3 is expressed in the lungs of mice exposed to several mixed Th2/Th17-polarizing allergic sensitization regimens. SAA instillation into the lungs elicits robust TLR2-, MyD88-, and IL-1-dependent pulmonary neutrophilic inflammation. Furthermore, SAA drives production of IL-1α, IL-1ß, IL-6, IL-23, and PGE(2), causes dendritic cell (DC) maturation, and requires TLR2, MyD88, and the NLRP3 inflammasome for secretion of IL-1ß by DCs and macrophages. CD4(+) T cells polyclonally stimulated in the presence of conditioned media from SAA-exposed DCs produced IL-17, and the capacity of polyclonally stimulated splenocytes to secrete IL-17 is dependent upon IL-1, TLR2, and the NLRP3 inflammasome. Additionally, in a model of allergic airway inflammation, administration of SAA to the lungs functions as an adjuvant to sensitize mice to inhaled OVA, resulting in leukocyte influx after Ag challenge and a predominance of IL-17 production from restimulated splenocytes that is dependent upon IL-1R signaling.


Assuntos
Alérgenos/fisiologia , Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/patologia , Proteína Amiloide A Sérica/fisiologia , Células Th17/imunologia , Células Th17/patologia , Alérgenos/genética , Animais , Proteínas de Transporte/genética , Polaridade Celular/genética , Polaridade Celular/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Inflamassomos/deficiência , Inflamassomos/genética , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Interleucina-1alfa/antagonistas & inibidores , Interleucina-1alfa/fisiologia , Interleucina-1beta/metabolismo , Interleucina-1beta/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Hipersensibilidade Respiratória/metabolismo , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Baço/imunologia , Baço/metabolismo , Baço/patologia , Células Th17/metabolismo , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/fisiologia
15.
J Immunol ; 186(7): 4140-6, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21368234

RESUMO

NKT cells are known to rapidly produce a large amount of cytokines upon activation. Although a number of signaling pathways that regulate the development of NKT cells have been identified, the signaling pathways involved in the regulation of NKT cell cytokine production remain unclear. In this study, we show that the p38 MAPK pathway is dispensable for the development of NKT cells. However, NKT cell cytokine production and NKT-mediated liver damage are highly dependent on activation of this pathway. p38 MAPK does not substantially affect cytokine gene expression in NKT cells, but it regulates the synthesis of cytokines through the Mnk-eIF4E pathway. Thus, in addition to gene expression, translational regulation by p38 MAPK could be a novel mechanism that contributes to the overall production of cytokine by NKT cells.


Assuntos
Citocinas/biossíntese , Citocinas/genética , Sistema de Sinalização das MAP Quinases/imunologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Modificação Traducional de Proteínas/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Hepatopatias/enzimologia , Hepatopatias/genética , Hepatopatias/imunologia , MAP Quinase Quinase 3/deficiência , MAP Quinase Quinase 3/genética , MAP Quinase Quinase 3/fisiologia , MAP Quinase Quinase 6/deficiência , MAP Quinase Quinase 6/genética , MAP Quinase Quinase 6/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células T Matadoras Naturais/enzimologia
16.
J Immunol ; 185(1): 144-56, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20530260

RESUMO

CD1d-restricted NKT cells make up an innate-like T cell subset that plays a role in amplifying the response of innate immune leukocytes to TLR ligands. The Slam locus contains genes that have been implicated in innate and adaptive immune responses. In this study, we demonstrate that divergent Slam locus haplotypes modulate the response of macrophages to the TLR4 ligand LPS through their control of NKT cell number and function. In response to LPS challenge in vivo, macrophage TNF production in Slam haplotype-2(+) 129S1/SvImJ and 129X1/SvJ mice was significantly impaired in comparison with macrophage TNF production in Slam haplotype-1(+) C57BL/6J mice. Although no cell-intrinsic differences in macrophage responses to LPS were observed between strains, 129 mice were found to be deficient in liver NKT cell number, in NKT cell cytokine production in response to the CD1d ligand alpha-galactosylceramide, and in NKT cell IFN-gamma production after LPS challenge in vivo. Using B6.129c1 congenic mice and adoptive transfer, we found that divergent Slam haplotypes controlled the response to LPS in vivo, as well as the diminished NKT cell number and function, and that these phenotypes were associated with differential expression of signaling lymphocytic activation molecule family receptors on NKT cells. These data suggest that the polymorphisms that distinguish two Slam haplotypes significantly modulate the innate immune response in vivo through their effect on NKT cells.


Assuntos
Antígenos CD/genética , Haplótipos , Imunidade Inata , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/fisiologia , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/imunologia , Receptores de Superfície Celular/genética , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Fígado/citologia , Fígado/imunologia , Fígado/metabolismo , Contagem de Linfócitos , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Baço/citologia , Baço/imunologia , Baço/metabolismo
17.
Am J Respir Cell Mol Biol ; 44(1): 11-23, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20118221

RESUMO

Indoleamine 2,3-dioxygenase (IDO) suppresses the functions of CD4(+) T cells through its ability to metabolize the essential amino acid tryptophan. Although the activity of IDO is required for the immunosuppression of allergic airway disease by the Toll-Like-Receptor 9 (TLR9) agonist, oligonucleotides comprised of cytosine and guanine nucleotides linked by phosphodiester bonds (CpG) DNA, it is unclear whether IDO expression by resident lung epithelial cells is sufficient to elicit these effects. Therefore, we created a transgenic mouse inducibly overexpressing IDO within nonciliated airway epithelial cells. Upon inhalation of formalin-fixed Aspergillus fumigatus hyphal antigens, the overexpression of IDO from airway epithelial cells of these mice reduced the number of CD4(+) T cells within the inflamed lung and impaired the capacity of antigen-specific splenic CD4(+) effector T cells to secrete the cytokines IL-4, IL-5, IL-13, and IFN-γ. Despite these effects, allergic airway disease pathology was largely unaffected in mice expressing IDO in airway epithelium. In support of the concept that dendritic cells are the major cell type contributing to the IDO-inducing effects of CpG DNA, mice expressing TLR9 only in the airway epithelium did not augment IDO expression subsequent to the administration of CpG DNA. Furthermore, the systemic depletion of CD11c(+) cells rendered mice incapable of CpG DNA-induced IDO expression. Our results demonstrate that an overexpression of IDO within the airway epithelium represents a novel mechanism by which the number of CD4(+) T cells recruited to the lung and their capacity to produce cytokines can be diminished in a model of allergic airway disease, and these results also highlight the critical role of dendritic cells in the antiasthmatic effects of IDO induction by CpG DNA.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Pulmão/enzimologia , Ativação Linfocitária , Aspergilose Pulmonar/enzimologia , Mucosa Respiratória/enzimologia , Animais , Antígenos de Fungos/imunologia , Aspergillus fumigatus/imunologia , Testes de Provocação Brônquica , Líquido da Lavagem Broncoalveolar/imunologia , Broncoconstrição , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/microbiologia , Linhagem Celular Transformada , Proliferação de Células , Técnicas de Cocultura , Citocinas/metabolismo , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Cinurenina/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/fisiopatologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oligodesoxirribonucleotídeos/farmacologia , Aspergilose Pulmonar/imunologia , Aspergilose Pulmonar/microbiologia , Aspergilose Pulmonar/fisiopatologia , Ratos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Regulação para Cima , Uteroglobina/genética , Uteroglobina/metabolismo
18.
J Immunol ; 182(6): 3728-34, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265151

RESUMO

The Lyme disease spirochete Borrelia burgdorferi is the only known human pathogen that directly activates invariant NKT (iNKT) cells. The number and activation kinetics of iNKT cells vary greatly among different strains of mice. We now report the role of the iNKT cell response in the pathogenesis of Lyme disease using C57BL/6 mice, a strain with optimal iNKT cell activation that is resistant to the development of spirochetal-induced inflammation. During experimental infection of B6 mice with B. burgdorferi, iNKT cells localize to the inflamed heart where they are activated by CD1d-expressing macrophages. Activation of iNKT cells in vivo results in the production of IFN-gamma, which we demonstrate ameliorates the severity of murine Lyme carditis by at least two mechanisms. First, IFN-gamma enhances the recognition of B. burgdorferi by macrophages, leading to increased phagocytosis of the spirochete. Second, IFN-gamma activation of macrophages increases the surface expression of CD1d, thereby facilitating further iNKT activation. Collectively, our data demonstrate that in the resistant background, B6, iNKT cells modulate the severity of murine Lyme carditis through the action of IFN-gamma, which appears to self-renew through a positive feedback loop during infection.


Assuntos
Interferon gama/biossíntese , Doença de Lyme/imunologia , Doença de Lyme/terapia , Miocardite/imunologia , Miocardite/terapia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Doença Aguda , Animais , Antígenos CD1d/biossíntese , Antígenos CD1d/genética , Antígenos CD1d/fisiologia , Borrelia burgdorferi/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Modelos Animais de Doenças , Retroalimentação Fisiológica/genética , Retroalimentação Fisiológica/imunologia , Interferon gama/fisiologia , Doença de Lyme/metabolismo , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/metabolismo , Células T Matadoras Naturais/patologia , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptor de Interferon gama
19.
Biomaterials ; 276: 121029, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34311317

RESUMO

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.


Assuntos
Bioprótese , Próteses Valvulares Cardíacas , Animais , Hipersensibilidade Alimentar , Galactosiltransferases/genética , Humanos , Pulmão , Suínos , Transplante Heterólogo
20.
J Exp Med ; 198(8): 1201-12, 2003 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-14568979

RESUMO

Natural killer cells constitute 50-90% of lymphocytes in human uterine decidua in early pregnancy. Here, CD56(bright) uterine decidual NK (dNK) cells were compared with the CD56(bright) and CD56(dim) peripheral NK cell subsets by microarray analysis, with verification of results by flow cytometry and RT-PCR. Among the approximately 10,000 genes studied, 278 genes showed at least a threefold change with P < or = 0.001 when comparing the dNK and peripheral NK cell subsets, most displaying increased expression in dNK cells. The largest number of these encoded surface proteins, including the unusual lectinlike receptors NKG2E and Ly-49L, several killer cell Ig-like receptors, the integrin subunits alpha(D), alpha(X), beta1, and beta5, and multiple tetraspanins (CD9, CD151, CD53, CD63, and TSPAN-5). Additionally, two secreted proteins, galectin-1 and progestagen-associated protein 14, known to have immunomodulatory functions, were selectively expressed in dNK cells.


Assuntos
Decídua/imunologia , Células Matadoras Naturais/classificação , Subpopulações de Linfócitos T/imunologia , Antígeno CD56/genética , Feminino , Galectina 1/metabolismo , Expressão Gênica , Glicodelina , Glicoproteínas/metabolismo , Proteínas de Homeodomínio , Humanos , Células Matadoras Naturais/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Proteínas da Gravidez/metabolismo , Receptores Imunológicos/metabolismo , Receptores de Células Matadoras Naturais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA