Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Toxicol Sci ; 176(2): 312-328, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32514536

ABSTRACT

Arsenic exposure via drinking water is a serious environmental health concern. Epidemiological studies suggest a strong association between prenatal arsenic exposure and subsequent childhood respiratory infections, as well as morbidity from respiratory diseases in adulthood, long after systemic clearance of arsenic. We investigated the impact of exclusive prenatal arsenic exposure on the inflammatory immune response and respiratory health after an adult influenza A virus (IAV) lung infection. C57BL/6J mice were exposed to 100 ppb sodium arsenite in utero, and subsequently infected with IAV (H1N1) after maturation to adulthood. Assessment of lung tissue and bronchoalveolar lavage fluid at various time points post-IAV infection reveals greater lung damage and inflammation in arsenic-exposed mice versus control mice. Single-cell RNA sequencing analysis of immune cells harvested from IAV-infected lungs suggests that the enhanced inflammatory response is mediated by dysregulation of innate immune function of monocyte-derived macrophages, neutrophils, natural killer cells, and alveolar macrophages. Our results suggest that prenatal arsenic exposure results in lasting effects on the adult host innate immune response to IAV infection, long after exposure to arsenic, leading to greater immunopathology. This study provides the first direct evidence that exclusive prenatal exposure to arsenic in drinking water causes predisposition to a hyperinflammatory response to IAV infection in adult mice, which is associated with significant lung damage.


Subject(s)
Arsenic , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/immunology , Prenatal Exposure Delayed Effects , Animals , Arsenic/adverse effects , Female , Gene Expression , Humans , Immunity, Innate , Influenza, Human , Lung , Mice , Mice, Inbred C57BL , Pregnancy , RNA-Seq , Single-Cell Analysis
2.
J Leukoc Biol ; 98(3): 423-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26019295

ABSTRACT

Elevated levels of solTNFR2 are observed in a variety of human pathophysiological conditions but regulation of TNFR2 levels during disease is not well understood. We found that solTNFR2 levels were increased following influenza infection or live-attenuated influenza virus challenge in mice and humans, respectively. As influenza-specific CD8(+) T cells up-regulated expression of TNFR2 after infection in mice, we hypothesized that CD8(+) T cells contributed, in part, to solTNFR2 production after influenza infection and were interested in the mechanisms by which CD8(+) T cells regulate TNFR2 shedding. Activation of these cells by TCR stimulation resulted in enhanced shedding of TNFR2 that required actin remodeling and lipid raft formation and was dependent on MAPK/ERK signaling. Furthermore, we identified ADAM17 as the protease responsible for TNFR2 shedding by CD8(+) T cells, with ADAM17 and TNFR2 required in "cis" for shedding to occur. We observed similar activation thresholds for TNF-α expression and TNFR2 shedding, suggesting that solTNFR2 functioned, in part, to regulate solTNF-α levels. Production of solTNFR2 by activated CD8(+) T cells reduced the availability of solTNF-α released by these cells, and TNFR2 blockade during influenza infection in mice enhanced the levels of solTNF-α, supporting this hypothesis. Taken together, this study identifies critical cellular mechanisms regulating TNFR2 shedding on CD8(+) T cells and demonstrates that TNFR2 contributes, in part, to the regulation of TNF-α levels during infection.


Subject(s)
ADAM Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , Influenza, Human/immunology , Influenza, Human/virology , Orthomyxoviridae Infections/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM17 Protein , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Influenza Vaccines/immunology , Mice, Inbred C57BL , Orthomyxoviridae Infections/virology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Solubility , Vaccines, Attenuated/immunology
3.
PLoS One ; 9(9): e108385, 2014.
Article in English | MEDLINE | ID: mdl-25251060

ABSTRACT

Virus infection triggers a CD8(+) T cell response that aids in virus clearance, but also expresses effector functions that may result in tissue injury. CD8(+) T cells express a variety of activating and inhibiting ligands, though regulation of the expression of inhibitory receptors is not well understood. The ligand for the inhibitory receptor, NKG2A, is the non-classical MHC-I molecule Qa1(b), which may also serve as a putative restricting element for the T cell receptors of purported regulatory CD8(+) T cells. We have previously shown that Qa1(b)-null mice suffer considerably enhanced immunopathologic lung injury in the context of CD8(+) T cell-mediated clearance of influenza infection, as well as evidence in a non-viral system that failure to ligate NKG2A on CD8(+) effector T cells may represent an important component of this process. In this report, we examine the requirements for induction of NKG2A expression, and show that NKG2A expression by CD8(+) T cells occurs as a result of migration from the MLN to the inflammatory lung environment, irrespective of peripheral antigen recognition. Further, we confirmed that NKG2A is a mediator in limiting immunopathology in virus infection using mice with a targeted deletion of NKG2A, and infecting the mutants with two different viruses, influenza and adenovirus. In neither infection is virus clearance altered. In influenza infection, the enhanced lung injury was associated with increased chemoattractant production, increased infiltration of inflammatory cells, and significantly enhanced alveolar hemorrhage. The primary mechanism of enhanced injury was the loss of negative regulation of CD8(+) T cell effector function. A similar effect was observed in the livers of mutant mice infected intravenously with adenovirus. These results demonstrate the immunoregulatory role of CD8(+) NKG2A expression in virus infection, which negatively regulates T cell effector functions and contributes to protection of tissue integrity during virus clearance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lung Injury/pathology , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Orthomyxoviridae Infections/immunology , Adenoviridae/pathogenicity , Adenoviridae Infections/immunology , Adenoviridae Infections/pathology , Adenoviridae Infections/veterinary , Adoptive Transfer , Animals , Gene Knockout Techniques , Influenza A virus/pathogenicity , Lung Injury/immunology , Lung Injury/virology , Mice , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/veterinary
4.
J Immunol ; 192(12): 5839-51, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24790150

ABSTRACT

TNF-α is a pleotropic cytokine that has both proinflammatory and anti-inflammatory functions during influenza infection. TNF-α is first expressed as a transmembrane protein that is proteolytically processed to release a soluble form. Transmembrane TNF-α (memTNF-α) and soluble TNF-α (solTNF-α) have been shown to exert distinct tissue-protective or tissue-pathologic effects in several disease models. However, the relative contributions of memTNF-α or solTNF-α in regulating pulmonary immunopathology following influenza infection are unclear. Therefore, we performed intranasal influenza infection in mice exclusively expressing noncleavable memTNF-α or lacking TNF-α entirely and examined the outcomes. We found that solTNF-α, but not memTNF-α, was required to limit the size of the immune response and the extent of injury. In the absence of solTNF-α, there was a significant increase in the CD8(+) T cell response, including virus-specific CD8(+) T cells, which was due in part to an increased resistance to activation-induced cell death. We found that solTNF-α mediates these immunoregulatory effects primarily through TNFR1, because mice deficient in TNFR1, but not TNFR2, exhibited dysregulated immune responses and exacerbated injury similar to that observed in mice lacking solTNF-α. We also found that solTNF-α expression was required early during infection to regulate the magnitude of the CD8(+) T cell response, indicating that early inflammatory events are critical for the regulation of the effector phase. Taken together, these findings suggest that processing of memTNF-α to release solTNF-α is a critical event regulating the immune response during influenza infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Membrane/immunology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Death/genetics , Cell Death/immunology , Cell Membrane/genetics , Cell Membrane/pathology , Influenza A Virus, H1N1 Subtype/genetics , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/immunology , Solubility , Tumor Necrosis Factor-alpha/genetics
5.
J Biol Chem ; 289(1): 152-62, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24247241

ABSTRACT

Cif (PA2934), a bacterial virulence factor secreted in outer membrane vesicles by Pseudomonas aeruginosa, increases the ubiquitination and lysosomal degradation of some, but not all, plasma membrane ATP-binding cassette transporters (ABC), including the cystic fibrosis transmembrane conductance regulator and P-glycoprotein. The goal of this study was to determine whether Cif enhances the ubiquitination and degradation of the transporter associated with antigen processing (TAP1 and TAP2), members of the ABC transporter family that play an essential role in antigen presentation and intracellular pathogen clearance. Cif selectively increased the amount of ubiquitinated TAP1 and increased its degradation in the proteasome of human airway epithelial cells. This effect of Cif was mediated by reducing USP10 deubiquitinating activity, resulting in increased polyubiquitination and proteasomal degradation of TAP1. The reduction in TAP1 abundance decreased peptide antigen translocation into the endoplasmic reticulum, an effect that resulted in reduced antigen available to MHC class I molecules for presentation at the plasma membrane of airway epithelial cells and recognition by CD8(+) T cells. Cif is the first bacterial factor identified that inhibits TAP function and MHC class I antigen presentation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antigen Presentation , Bacterial Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Proteasome Endopeptidase Complex/metabolism , Pseudomonas aeruginosa/metabolism , Ubiquitination , Virulence Factors/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Protein Transport/genetics , Protein Transport/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/immunology , Ubiquitin Thiolesterase/metabolism , Virulence Factors/genetics , Virulence Factors/immunology
6.
PLoS One ; 8(11): e79340, 2013.
Article in English | MEDLINE | ID: mdl-24223177

ABSTRACT

Influenza infection in humans evokes a potent CD8(+) T-cell response, which is important for clearance of the virus but may also exacerbate pulmonary pathology. We have previously shown in mice that CD8(+) T-cell expression of TNF-α is required for severe and lethal lung injury following recognition of an influenza antigen expressed by alveolar epithelial cells. Since TNF-α is first expressed as a transmembrane protein that is then proteolytically processed to release a soluble form, we sought to characterize the role of TNF-α processing in CD8(+) T-cell-mediated injury. In this study we observed that inhibition of ADAM17-mediated processing of TNF-α by CD8(+) T cells significantly attenuated the diffuse alveolar damage that occurs after T-cell transfer, resulting in enhanced survival. This was due in part to diminished chemokine expression, as TNF-α processing was required for lung epithelial cell expression of CXCL2 and the subsequent inflammatory infiltration. We confirmed the importance of CXCL2 expression in acute lung injury by transferring influenza-specific CD8(+) T cells into transgenic mice lacking CXCR2. These mice exhibited reduced airway infiltration, attenuated lung injury, and enhanced survival. Theses studies describe a critical role for TNF-α processing by CD8(+) T cells in the initiation and severity of acute lung injury, which may have important implications for limiting immunopathology during influenza infection and other human infectious or inflammatory diseases.


Subject(s)
ADAM Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation , Lung Injury/immunology , Lung Injury/virology , Proteolysis , Tumor Necrosis Factor-alpha/metabolism , ADAM Proteins/deficiency , ADAM17 Protein , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Lung Injury/metabolism , Lung Injury/pathology , Macrophages/immunology , Mice , Neutrophil Infiltration , Receptors, Interleukin-8B/metabolism , Signal Transduction , Solubility , Tumor Necrosis Factor-alpha/chemistry
7.
Environ Health Perspect ; 117(9): 1441-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19750111

ABSTRACT

BACKGROUND: Arsenic exposure is a significant worldwide environmental health concern. We recently reported that 5-week exposure to environmentally relevant levels (10 and 100 ppb) of As in drinking water significantly altered components of the innate immune response in mouse lung, which we hypothesize is an important contributor to the increased risk of lung disease in exposed human populations. OBJECTIVES: We investigated the effects of As exposure on respiratory influenza A (H1N1) virus infection, a common and potentially fatal disease. METHODS: In this study, we exposed C57BL/6J mice to 100 ppb As in drinking water for 5 weeks, followed by intranasal inoculation with a sub lethal dose of influenza A/PuertoRico/8/34 (H1N1) virus. Multiple end points were assessed postinfection. RESULTS: Arsenic was associated with a number of significant changes in response to influenza, including an increase in morbidity and higher pulmonary influenza virus titers on day 7 post-infection. We also found many alterations in the immune response relative to As-unexposed controls, including a decrease in the number of dendritic cells in the mediastinal lymph nodes early in the course of infection. CONCLUSIONS: Our data indicate that chronic As exposure significantly compromises the immune response to infection. Alterations in response to repeated lung infection may also contribute to other chronic illnesses, such as bronchiectasis, which is elevated by As exposure in epidemiology studies.


Subject(s)
Arsenic/toxicity , Influenza A virus/isolation & purification , Orthomyxoviridae Infections/immunology , Animals , Bronchoalveolar Lavage Fluid , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL
8.
J Immunol ; 179(6): 3535-42, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785787

ABSTRACT

Increasing age is associated with the development of CD8+ T cell clonal expansions (TCE) that can dominate the peripheral T cell repertoire and interfere with immune responses to infection and vaccination. Some TCE are driven by chronic infections, consistent with dysregulated outgrowth of T cell clones in response to persistent antigenic stimulation. However, a second class of TCE develops with age in the absence of chronic infections and is poorly understood in terms of origin or Ag dependence. In this study, we present evidence that Ag-specific TCE develop at high frequencies from conventional memory CD8+ T cell pools elicited by nonpersistent influenza and parainfluenza virus infections. Putative TCE occurred in both the central- and effector-memory CD8+ T cell populations and did not require Ag for their maintenance. In addition, they were similar to normal memory T cells in terms of phenotype and function, suggesting that they develop stochastically from the memory T cell pool. These data suggest that memory T cell pools become progressively dysregulated over time and this may have a significant impact on immune responsiveness in the aged.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza A Virus, H3N2 Subtype/immunology , Lymphocyte Activation/immunology , Respiratory Tract Infections/immunology , Sendai virus/immunology , T-Lymphocyte Subsets/immunology , Acute Disease , Aging/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Clone Cells , Epitopes, T-Lymphocyte/biosynthesis , Female , Lymphocyte Count , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Respiratory Tract Infections/pathology , Respirovirus Infections/immunology , Respirovirus Infections/pathology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology
9.
Exp Gerontol ; 42(5): 427-31, 2007 May.
Article in English | MEDLINE | ID: mdl-17197143

ABSTRACT

The capacity of the immune system to mediate effective immune responses to pathogens declines with age. In the case of immune responses to newly encountered antigens, several studies have demonstrated that this decline reflects both a loss of naïve T cells and changes in the repertoire and function of these cells over time. However, comparatively little is known about the impact of age on established memory T cells pools. Here we discuss age-related changes in memory CD8(+) T cell pools elicited by influenza and parainfluenza viruses and the impact of these changes on immunity in general.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Virus Diseases/immunology , Humans , Immunologic Memory
10.
Immunol Rev ; 211: 119-32, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16824122

ABSTRACT

The respiratory tract is characterized by its large surface area and the close association of an extensive vasculature with the external environment. As such, the respiratory tract is a major portal of entry for many pathogens. The immune system is able to effectively control most pulmonary pathogens and establish immunological memory that is capable of mediating an accelerated and enhanced recall response to secondary pathogen challenge. A key component of the recall response in the lung involves the rapid response of antigen-specific memory CD8+ T cells. Recent studies have shown that memory CD8+ T cells are extremely heterogeneous in terms of phenotype, function, anatomical distribution, and longevity. However, we have little understanding of how the different subsets of memory cells actually contribute to the recall response, especially with respect to peripheral or mucosal sites, such as the lung. Since immunological memory is the cornerstone of vaccination, it is essential that we understand how different memory CD8+ T-cell subsets are initially generated, maintained over time, and contribute to recall responses. This review focuses on memory T cells that mediate recall responses to influenza and parainfluenza virus infections in the lung.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Respiratory Tract Infections/immunology , Animals , Humans , Mice , Respiratory Tract Infections/virology , Vaccination
11.
J Immunol ; 176(1): 537-43, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365448

ABSTRACT

Effector memory T cell populations in the periphery play a key role in cellular immune responses to secondary infections. However, it is unclear how these populations are maintained under steady-state conditions in nonlymphoid peripheral sites, such as the lung airways. In this study, we show that LFA-1 expression is selectively down-regulated following entry of memory T cells into the lung airways. Using Sendai virus as a mouse model of respiratory virus infection, we use LFA-1 expression levels to demonstrate that effector memory T cell populations in the lung airways are maintained by continual recruitment of new cells from the circulation. The rate of memory cell recruitment is surprisingly rapid, resulting in replacement of 90% of the population every 10 days, and is maintained for well over 1 year following viral clearance. These data indicate that peripheral T cell memory is dynamic and depends on a systemic source of T cells.


Subject(s)
Immunologic Memory , Lung/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Bronchoalveolar Lavage Fluid/cytology , CD11a Antigen/immunology , CD11a Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte , Lung/microbiology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Respirovirus Infections/immunology , Sendai virus/immunology
12.
J Exp Med ; 202(1): 123-33, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15983064

ABSTRACT

Although the absolute number of memory CD8+ T cells established in the spleen following antigen encounter remains stable for many years, the relative capacity of these cells to mediate recall responses is not known. Here we used a dual adoptive transfer approach to demonstrate a progressive increase in the quality of memory T cell pools in terms of their ability to proliferate and accumulate at effector sites in response to secondary pathogen challenge. This temporal increase in efficacy occurred in CD62L lo (effector memory) and CD62L hi (central memory) subpopulations, but was most prominent in the CD62L hi subpopulation. These data indicate that the contribution of effector memory and central memory T cells to the recall response changes substantially over time.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Aging/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , L-Selectin/metabolism , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Respirovirus Infections/immunology , Sendai virus , T-Lymphocyte Subsets/cytology , Time Factors
13.
Gastroenterology ; 127(1): 119-26, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15236178

ABSTRACT

BACKGROUND & AIMS: The loss of homeostasis is a hallmark of inflammatory bowel disease. Oral infection of susceptible mice with Toxoplasma gondii results in an acute lethal ileitis characterized by increased interferon gamma, tumor necrosis factor alpha, and inducible nitric oxide synthase; homeostasis results from transforming growth factor beta production by intraepithelial lymphocytes. The synthetic oleanane triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) is a potent anti-inflammatory molecule previously shown in vitro to suppress the de novo synthesis of inducible nitric oxide synthase and to induce the transcription and activation of genes from the transforming growth factor beta signaling pathway. METHODS: We evaluated the immune response in the small intestine and by intraepithelial lymphocytes after a single intraperitoneal dose of CDDO at the time of T. gondii oral infection. We abrogated the homeostatic effects of CDDO by blocking transforming growth factor beta in vivo. RESULTS: CDDO acid prevented ileitis development through the global down-regulation of inflammatory cytokines and chemokines. Total transforming growth factor beta(1) production by the intraepithelial lymphocytes increased, as did Smad2 expression. Blocking transforming growth factor beta reversed CDDO-induced protection and prevented the up-regulation of Smad2 in the small intestine. CONCLUSIONS: CDDO acid is a novel anti-inflammatory molecule capable of preventing ileitis by activating the transforming growth factor beta signaling pathway in a pathogen-driven ileitis model. This could represent a new treatment of inflammatory bowel disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Ileitis/parasitology , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/administration & dosage , Toxoplasmosis/complications , Transforming Growth Factor beta/drug effects , Animals , Cytokines/drug effects , Cytokines/immunology , Down-Regulation/genetics , Down-Regulation/immunology , Female , Ileitis/prevention & control , Injections, Intraperitoneal , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Models, Animal , Signal Transduction/immunology , Transforming Growth Factor beta/physiology
14.
Proc Natl Acad Sci U S A ; 101(9): 3005-10, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14976252

ABSTRACT

Fibrinogen-like protein 2 (Fgl2, fibroleukin) is a leukocyte product that exhibits significant homology to secreted proteins of diverse function, including growth factors, lectins, and components of extracellular matrix. Prior studies found that Fgl2 is IFN gamma-inducible, possesses direct coagulant activity, and inhibits T cell proliferation and dendritic cell maturation in vitro. Here, we demonstrate that Fgl2 expression is up-regulated during type 1 immunity in vivo and establish that such up-regulation is IFN gamma-, signal transducer and activation of transcription protein 1-, and IFN response factor 1-dependent. To investigate functional roles for Fgl2 during type 1 immunity, we generated Fgl2-deficient mice. Those animals are born at predicted Mendelian frequencies, appear overtly healthy, and contain normal numbers and frequencies of lymphoid cells. Although Fgl2 is IFN gamma-inducible and putatively regulates T cell activation/proliferation, we demonstrate that Fgl2-deficient and control mice exhibit similar degrees of T cell expansion, immunopathology, and/or pathogen burdens during protozoan (Toxoplasma gondii), bacterial (Yersinia enterocolitica, Listeria monocytogenes, and Mycobacterium tuberculosis), and viral (murine gamma-herpesvirus-68 and Sendai) infections. Fgl2-deficient mice also reject allografts with similar kinetics as control mice. Moreover, despite prior reports that Fgl2 functions as a procoagulant enzyme, we demonstrate that Fgl2-deficient and control mice produce similar levels of fibrin, a product of the coagulation cascade, during T. gondii infection and allograft rejection. Together, our findings suggest that Fgl2, although highly conserved and IFN gamma-inducible, is not a critical mediator of either type 1 immunity or immune-associated coagulant activity.


Subject(s)
Interferon-gamma/immunology , T-Lymphocytes/immunology , Animals , Bacterial Infections/immunology , Blood Coagulation/immunology , DNA Primers , Disease Models, Animal , Fibrin/immunology , Heart Transplantation/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Toxoplasmosis/immunology , Transplantation, Homologous , Virus Diseases/immunology
15.
J Immunol ; 171(7): 3338-42, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14500625

ABSTRACT

Previous studies have shown that long-lived memory CD8(+) T cells persist in the lung airways following the resolution of a murine Sendai virus infection. These cells are CD11a(low), noncytolytic, and do not proliferate in the lung airways raising the possibility that they are "end stage" or terminally differentiated memory cells. In this current report, we investigated the functional characteristics of these cells by analyzing their capacity to respond to secondary viral infection outside of the lung environment. We show that, after transfer into the bloodstream, CD11a(low) memory T cells from the lung airways can return to the secondary lymphoid tissue and respond to a secondary viral challenge. Furthermore, these cells re-express CD11a, which may contribute to their migratory and proliferative capacity. These data demonstrate that lung airway memory CD8(+) T cells are not terminally differentiated cells and retain the capacity to mediate recall responses to infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lung/cytology , Lung/immunology , T-Lymphocyte Subsets/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD11a Antigen/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/virology , Cell Division/immunology , Cell Movement/immunology , Female , Injections, Intravenous , Lung/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Respirovirus Infections/immunology , Respirovirus Infections/pathology , Sendai virus/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , T-Lymphocyte Subsets/virology
16.
J Exp Med ; 198(3): 399-410, 2003 Aug 04.
Article in English | MEDLINE | ID: mdl-12885871

ABSTRACT

The specificity of CD8+ T cell responses can vary dramatically between primary and secondary infections. For example, NP366-374/Db- and PA224-233/Db-specific CD8+ T cells respond in approximately equal numbers to a primary influenza virus infection in C57BL/6 mice, whereas NP366-374/Db-specific CD8+ T cells dominate the secondary response. To investigate the mechanisms underlying this changing pattern of immunodominance, we analyzed the role of antigen presentation in regulating the specificity of the T cell response. The data show that both dendritic and nondendritic cells are able to present the NP366-374/Db epitope, whereas only dendritic cells effectively present the PA224-233/Db epitope after influenza virus infection, both in vitro and in vivo. This difference in epitope expression favored the activation and expansion of NP366-374/Db-specific CD8+ memory T cells during secondary infection. The data also show that the immune response to influenza virus infection may involve T cells specific for epitopes, such as PA224-233/Db, that are poorly expressed at the site of infection. In this regard, vaccination with the PA224-233 peptide actually had a detrimental effect on the clearance of a subsequent influenza virus infection. Thus, differential antigen presentation impacts both the specificity of the T cell response and the efficacy of peptide-based vaccination strategies.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Immunodominant Epitopes/immunology , Immunologic Memory , Orthomyxoviridae Infections/immunology , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes , Female , Mice , Mice, Inbred C57BL , Orthomyxoviridae/immunology , Orthomyxoviridae/metabolism , Peptide Fragments/immunology , Viral Core Proteins/immunology
17.
J Immunol ; 170(3): 1423-9, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12538703

ABSTRACT

Previous studies have shown that heterologous viral infections have a significant impact on pre-existing memory T cell populations in secondary lymphoid organs through a combination of cross-reactive and bystander effects. However, the impact of heterologous viral infections on effector/memory T cells in peripheral sites is not well understood. In this study, we have analyzed the impact of a heterologous influenza virus infection on Sendai virus-specific CD8(+) effector/memory cells present in the lung airways. The data show a transient increase in the numbers of Sendai virus nucleoprotein 324-332/K(b)-specific CD8(+) memory T cells in the airways of the influenza-infected mice peaking around day 4 postinfection. Intratracheal transfer studies and 5-bromo-2'-deoxyuridine incorporation demonstrate that this increase is due to the recruitment of resting memory cells into the airways. In addition, the data show that these immigrating memory cells are phenotypically distinct from the resident memory T cells of the lung airways. A similar influx of nonproliferating Sendai virus nucleoprotein 324-332/K(b)-specific CD8(+) memory T cells is also induced by a secondary (homologous) infection with Sendai virus. Together, these data suggest that inflammation can accelerate memory T cell migration to nonlymphoid tissues and is a part of the normal recall response during respiratory infections.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Epitopes, T-Lymphocyte/immunology , Immunologic Memory , Lung/immunology , Orthomyxoviridae Infections/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Cell Division/immunology , Female , Immunophenotyping , Influenza A virus/immunology , Lung/cytology , Lung/virology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/virology , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Up-Regulation/immunology , Vaccinia/immunology , Vaccinia/virology , Vaccinia virus/immunology
18.
J Immunol ; 169(9): 4976-81, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391211

ABSTRACT

Recent studies have shown that virus-specific effector memory T cells can be recovered from the lung airways long after clearance of a respiratory virus infection. These cells are thought to play an important role in the recall response to secondary viral infection. It is currently unclear whether these cells actually persist at this site or are maintained by continual proliferation and recruitment. In this study, we have analyzed the mechanisms underlying the persistence of memory CD8(+) T cells in the lung airway lumina following recovery from a respiratory virus infection. The data identify two distinct populations of memory cells. First, a large population Ag-specific CD8(+) T cells is deposited in the airways during the acute response to the virus. These cells persist in a functional state for several weeks with minimal further division. Second, a smaller population of Ag-specific CD8(+) T cells is maintained in the lung airways by homeostatic proliferation and migration to lung airways after viral clearance. This rate of proliferation is identical to that observed in the spleen, suggesting that these cells may be recent immigrants from the lymphoid organs. These data have significant implications for vaccines designed to promote cellular immunity at mucosal sites such as the lung.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunologic Memory , Lung/cytology , Lung/immunology , Sendai virus/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Division/immunology , Cell Survival/immunology , Convalescence , Female , Lung/virology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins , Nucleoproteins/immunology , Respirovirus Infections/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , T-Lymphocytes, Regulatory/virology , Viral Core Proteins/immunology
19.
Microbes Infect ; 4(10): 1091-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12191659

ABSTRACT

Recent studies have identified distinct populations of memory T cells that persist in the lungs following respiratory virus infections, and contribute to the control of secondary virus infections. Here we discuss the establishment, maintenance and recall of memory T cells in the lung.


Subject(s)
Immunologic Memory/immunology , Lung Diseases/immunology , Lung Diseases/virology , T-Lymphocytes/immunology , Humans , Immunophenotyping , Lymphoid Tissue/immunology , Respiratory System/immunology , Viral Vaccines/immunology
20.
Infect Immun ; 70(2): 434-43, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11796568

ABSTRACT

T-cell immunity is critical for survival of hosts infected with Toxoplasma gondii. Among the cells in the T-cell population, CD8(+) T cells are considered the major effector cells against this parasite. It is believed that CD4(+) T cells may be crucial for induction of the CD8(+)-T-cell response against T. gondii. In the present study, CD4(-/-) mice were used to evaluate the role of conventional CD4(+) T cells in the immune response against T. gondii infection. CD4(-/-) mice infected with T. gondii exhibited lower gamma interferon (IFN-gamma) messages in the majority of their tissues. As a result, mortality due to a hyperinflammatory response was prevented in these animals. Interestingly, T. gondii infection induced a normal antigen-specific CD8(+)-T-cell immune response in CD4(-/-) mice. No difference in generation of precursor cytotoxic T lymphocytes (pCTL) or in IFN-gamma production by the CD8(+)-T-cell populations from the knockout and wild-type animals was observed. However, the mutant mice were not able to sustain CD8(+)-T-cell immunity. At 180 days after infection, the CD8(+)-T-cell response in the knockout mice was depressed, as determined by pCTL and IFN-gamma assays. Loss of CD8(+)-T-cell immunity at this time was confirmed by adoptive transfer experiments. Purified CD8(+) T cells from CD4(-/-) donors that had been immunized 180 days earlier failed to protect the recipient mice against a lethal infection. Our study demonstrated that although CD8(+)-T-cell immunity can be induced in the absence of conventional CD4(+) T cells, it cannot be maintained without such cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Toxoplasmosis/immunology , Administration, Oral , Animals , Interferon-gamma/biosynthesis , Intestine, Small/pathology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Cytotoxic/immunology , Toxoplasma/immunology , Toxoplasmosis/parasitology , Toxoplasmosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...