Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell ; 175(6): 1634-1650.e17, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30433869

ABSTRACT

Innate immune memory is an emerging area of research. However, innate immune memory at major mucosal sites remains poorly understood. Here, we show that respiratory viral infection induces long-lasting memory alveolar macrophages (AMs). Memory AMs are programed to express high MHC II, a defense-ready gene signature, and increased glycolytic metabolism, and produce, upon re-stimulation, neutrophil chemokines. Using a multitude of approaches, we reveal that the priming, but not maintenance, of memory AMs requires the help from effector CD8 T cells. T cells jump-start this process via IFN-γ production. We further find that formation and maintenance of memory AMs are independent of monocytes or bone marrow progenitors. Finally, we demonstrate that memory AMs are poised for robust trained immunity against bacterial infection in the lung via rapid induction of chemokines and neutrophilia. Our study thus establishes a new paradigm of immunological memory formation whereby adaptive T-lymphocytes render innate memory of mucosal-associated macrophages.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Innate , Lung/immunology , Macrophages, Alveolar/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Immunologic Memory , Lung/cytology , Macrophages, Alveolar/cytology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Monocytes/cytology , Monocytes/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , T-Lymphocytes, Helper-Inducer/cytology
2.
Vaccine ; 36(37): 5636-5644, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30104115

ABSTRACT

The first evidence in humans that a safe and effective preventive vaccine for HIV is possible came from the phase III HIV clinical trial RV144 in Thailand. This trial was based on a prime/boost combination of a recombinant canarypox vaccine and two glycoprotein 120 proteins (ALVAC-HIV and AIDSVAX B/E). A pivotal phase IIb/III trial has recently commenced in the Republic of South Africa, for which the infectious titer assay was applied as the quantitative release test for the ALVAC-HIV vaccine. The infectious titer assay measures the ability of the vaccine vector to infect target permissive cells, but does not indicate if the vaccine transgenes are expressed. We have developed a high-throughput biological activity assay that provides results in agreement with the infectious titer assay. This assay uses flow cytometry to quantify expression of ALVAC-HIV encoded proteins gp120 and p24 in human cells. This transgene expression is detected by two cross-clade-reactive, biologically functional human anti-gp120 monoclonal antibodies isolated from clinical trial participants and a commercial mouse anti-p24 monoclonal antibody. The relative biological activity of the vaccine test sample is calculated by comparison of the test sample dose-response curve against that of a reference standard. We show that the novel biological activity assay is specific, accurate, precise, stability-indicating, and robust. The assay is being used for characterization of ALVAC-HIV (vCP2438) product, the efficacy of which is being evaluated in the pivotal phase IIb/III clinical trial HVTN702. The biological activity assay has the potential to indicate vaccine consistency and quality as a complement to the infectious titer assay.


Subject(s)
AIDS Vaccines/immunology , Flow Cytometry , HIV Antibodies/immunology , High-Throughput Screening Assays , AIDS Vaccines/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , HIV Core Protein p24/immunology , HIV Envelope Protein gp120/immunology , HeLa Cells , Humans , Jurkat Cells , Sensitivity and Specificity
3.
J Immunol ; 199(7): 2555-2569, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28827285

ABSTRACT

Although most novel tuberculosis (TB) vaccines are designed for delivery via the muscle or skin for enhanced protection in the lung, it has remained poorly understood whether systemic vaccine-induced memory T cells can readily home to the lung mucosa prior to and shortly after pathogen exposure. We have investigated this issue by using a model of parenteral TB immunization and intravascular immunostaining. We find that systemically induced memory T cells are restricted to the blood vessels in the lung, unable to populate either the lung parenchymal tissue or the airway under homeostatic conditions. We further find that after pulmonary TB infection, it still takes many days before such T cells can enter the lung parenchymal tissue and airway. We have identified the acquisition of CXCR3 expression by circulating T cells to be critical for their entry to these lung mucosal compartments. Our findings offer new insights into mucosal T cell biology and have important implications in vaccine strategies against pulmonary TB and other intracellular infections in the lung.


Subject(s)
Lung/immunology , Mycobacterium tuberculosis/immunology , Receptors, CXCR3/metabolism , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Adoptive Transfer , Animals , Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Immunization , Immunologic Memory , Leukocytes/immunology , Lung/cytology , Lung/microbiology , Mice , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Signal Transduction , Tuberculosis Vaccines/administration & dosage , Tuberculosis, Pulmonary/microbiology
4.
J Infect Dis ; 214(12): 1996-2005, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27703038

ABSTRACT

BACKGROUND: Whether a candidate tuberculosis vaccine induces clinically relevant protective T-cell repertoires in humans will not be known until the completion of costly efficacy clinical trials. METHODS: We have developed an integrated immunologic approach to investigate the clinical relevance of T cells induced by a novel tuberculosis vaccine in a phase 1 trial. This approach consists of screening for likely dominant T-cell epitopes, establishing antigen-specific memory T-cell lines for identifying CD8+ and CD4+ T-cell epitopes, determining the ability of vaccine-induced T cells to inhibit mycobacterial growth in infected cells, and examining the genetic diversity of HLA recognition and the clinical relevance of identified T-cell epitopes. RESULTS: A single-dose immunization in BCG-primed adults with an adenovirus-based tuberculosis vaccine elicits a repertoire of memory T cells capable of recognizing multiple Ag85A epitopes. These T cells are polyfunctional and cytotoxic and can inhibit mycobacterial growth in infected target cells. Some identified T-cell epitopes are promiscuous and recognizable by the common HLA alleles. These epitopes are clinically relevant to the epitopes identified in people with latent Mycobacterium tuberculosis infection and treated patients with tuberculosis. CONCLUSIONS: These data support further clinical development of this candidate vaccine. Our approach helps fill the gap in clinical tuberculosis vaccine development.


Subject(s)
Adenoviridae/genetics , Drug Carriers , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Acyltransferases/genetics , Acyltransferases/immunology , Adult , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Mycobacterium tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
5.
J Immunol ; 195(6): 2900-7, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26268652

ABSTRACT

Emerging evidence suggests a role of B cells in host defense against primary pulmonary tuberculosis (TB). However, the role of B cells in TB vaccine-induced protective T cell immunity still remains unknown. Using a viral-vectored model TB vaccine and a number of experimental approaches, we have investigated the role of B cells in respiratory mucosal vaccine-induced T cell responses and protection against pulmonary TB. We found that respiratory mucosal vaccination activated Ag-specific B cell responses. Whereas respiratory mucosal vaccination elicited Ag-specific T cell responses in the airway and lung interstitium of genetic B cell-deficient (Jh(-/-) knockout [KO]) mice, the levels of airway T cell responses were lower than in wild-type hosts, which were associated with suboptimal protection against pulmonary Mycobacterium tuberculosis challenge. However, mucosal vaccination induced T cell responses in the airway and lung interstitium and protection in B cell-depleted wild-type mice to a similar extent as in B cell-competent hosts. Furthermore, by using an adoptive cell transfer approach, reconstitution of B cells in vaccinated Jh(-/-) KO mice did not enhance anti-TB protection. Moreover, respiratory mucosal vaccine-activated T cells alone were able to enhance anti-TB protection in SCID mice, and the transfer of vaccine-primed B cells alongside T cells did not further enhance such protection. Alternatively, adoptively transferring vaccine-primed T cells from Jh(-/-) KO mice into SCID mice only provided suboptimal protection. These data together suggest that B cells play a minimal role, and highlight a central role by T cells, in respiratory mucosal vaccine-induced protective immunity against M. tuberculosis.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Adoptive Transfer , Animals , B-Lymphocytes/transplantation , Female , Immunity, Mucosal/immunology , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Mycobacterium tuberculosis/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Tuberculosis, Pulmonary/prevention & control , Vaccination
6.
PLoS One ; 10(6): e0131175, 2015.
Article in English | MEDLINE | ID: mdl-26098423

ABSTRACT

Tuberculosis (TB) remains a global pandemic despite the use of Bacillus Calmette-Guérin (BCG) vaccine, partly because BCG fails to effectively control adult pulmonary TB. The introduction of novel boost vaccines such as the human Adenovirus 5-vectored AdHu5Ag85A could improve and prolong the protective immunity of BCG immunization. Age at which BCG immunization is implemented varies greatly worldwide, and research is ongoing to discover the optimal stage during childhood to administer the vaccine, as well as when to boost the immune response with potential novel vaccines. Using a murine model of subcutaneous BCG immunization followed by intranasal AdHu5Ag85A boosting, we investigated the impact of age at BCG immunization on protective efficacy of BCG prime and AdHu5Ag85A boost immunization-mediated protection. Our results showed that age at parenteral BCG priming has limited impact on the efficacy of BCG prime-AdHu5Ag85A respiratory mucosal boost immunization-enhanced protection. However, when BCG immunization was delayed until the maturity of the immune system, longer sustained memory T cells were generated and resulted in enhanced boosting effect on T cells of AdHu5Ag85A respiratory mucosal immunization. Our findings hold implications for the design of new TB immunization protocols for humans.


Subject(s)
BCG Vaccine/pharmacology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/therapeutic use , Tuberculosis, Pulmonary/prevention & control , Age Factors , Animals , BCG Vaccine/immunology , Disease Models, Animal , Female , Flow Cytometry , Immunologic Memory , Male , Mice , Mice, Inbred BALB C , Respiratory Mucosa/immunology , T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology
7.
Eur J Immunol ; 44(5): 1375-86, 2014 May.
Article in English | MEDLINE | ID: mdl-24519467

ABSTRACT

The immune mechanisms underlying delayed induction of Th1-type immunity in the lungs following pulmonary mycobacterial infection remain poorly understood. We have herein investigated the underlying immune mechanisms for such delayed responses and whether a selected innate immune-modulating strategy can accelerate Th1-type responses. We have found that, in the early stage of pulmonary infection with attenuated Mycobacterium tuberculosis (M.tb H37Ra), the levels of infection in the lung continue to increase logarithmically until days 14 and 21 postinfection in C57BL/6 mice. The activation of innate immune responses, particularly DCs, in the lung is delayed. This results in a delay in the subsequent downstream immune responses including the migration of antigen-bearing DCs to the draining lymph node (dLN), the Th1-cell priming in dLN, and the recruitment of Th1 cells to the lung. However, single lung mucosal exposure to the TLR agonist FimH postinfection is able to accelerate protective Th1-type immunity via facilitating DC migration to the lung and draining lymph nodes, enhancing DC antigen presentation and Th1-cell priming. These findings hold implications for the development of immunotherapeutic and vaccination strategies and suggest that enhancement of early innate immune activation is a viable option for improving Th1-type immunity against pulmonary mycobacterial diseases.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Lung/immunology , Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Tuberculosis, Pulmonary/immunology , Animals , Dendritic Cells/microbiology , Dendritic Cells/pathology , Lung/microbiology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Mice , Mice, Transgenic , Th1 Cells/pathology , Time Factors , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/pathology
8.
Article in English | MEDLINE | ID: mdl-26015944

ABSTRACT

Pneumococcal infections are the leading cause of community-acquired pneumonia. Although the type 1 interferon-α (IFN-α) is a well-known antiviral cytokine, the role of IFN-α in antipneumococcal host defense and its therapeutic potential remain poorly understood. We have investigated these issues by using a murine transgene expression model. We found that in control animals, Streptococcus pneumoniae infection caused severe weight loss and excessive lung inflammation, associated with rapid bacterial outgrowth. In contrast, the animals that received a single dose of an adenoviral vector expressing IFN-α prior to pneumococcal infection demonstrated rapid and effective control of bacterial replication and lung inflammation and improved clinical outcome. Enhanced protection by IFN-α was due to increased activation of neutrophils and macrophages with increased release of reactive oxygen and nitrogen species and bacterial killing. Furthermore, we found that raised levels of IFN-α in the lung remained immune protective even when the gene transfer vector was given at a time postpneumococcal infection. Our study thus shows that the classically antiviral type 1 IFN can be exploited for enhancing immunity against pneumococcal infection via its activating effects on innate immune cells. Our findings hold implications for the therapeutic use of IFN-α gene transfer strategies to combat pneumococcal infections.

9.
Sci Transl Med ; 5(205): 205ra134, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24089406

ABSTRACT

There is an urgent need to develop new tuberculosis (TB) vaccines to safely and effectively boost Bacille Calmette-Guérin (BCG)-triggered T cell immunity in humans. AdHu5Ag85A is a recombinant human type 5 adenovirus (AdHu5)-based TB vaccine with demonstrated efficacy in a number of animal species, yet it remains to be translated to human applications. In this phase 1 study, we evaluated the safety and immunogenicity of AdHu5Ag85A in both BCG-naïve and previously BCG-immunized healthy adults. Intramuscular immunization of AdHu5Ag85A was safe and well tolerated in both trial volunteer groups. Moreover, although AdHu5Ag85A was immunogenic in both trial volunteer groups, it much more potently boosted polyfunctional CD4(+) and CD8(+) T cell immunity in previously BCG-vaccinated volunteers. Furthermore, despite prevalent preexisting anti-AdHu5 humoral immunity in most of the trial volunteers, we found little evidence that such preexisting anti-AdHu5 immunity significantly dampened the potency of AdHu5Ag85A vaccine. This study supports further clinical investigations of the AdHu5Ag85A vaccine for human applications. It also suggests that the widely perceived negative effect of preexisting anti-AdHu5 immunity may not be universally applied to all AdHu5-based vaccines against different types of human pathogens.


Subject(s)
Adenoviruses, Human/immunology , Immunity/immunology , T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Healthy Volunteers , Humans , Interferon-gamma/metabolism , Male , Middle Aged , T-Lymphocytes/metabolism , Tuberculosis Vaccines/adverse effects , Vaccination/adverse effects , Young Adult
10.
Am J Pathol ; 183(3): 868-80, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23831294

ABSTRACT

Bacterial superinfection and associated lung immunopathology are major contributors to hospitalizations and mortality after influenza. However, the underlying mechanisms and effective intervention strategies remain poorly defined. By using a model of influenza and pneumococcal superinfection, we found that dual-infected animals experienced rapid weight loss and succumbed to infection. Bacterial outgrowth, dysregulated cytokines, including keratinocyte-derived chemokine and macrophage inflammatory protein 2, and severe lung neutrophilia and immunopathology were linked to the poor clinical outcome. In vivo neutralization of highly induced macrophage inflammatory protein 2 did not affect clinical outcome, bacterial loads, or lung immunopathology. On the other hand, in vivo neutrophil depletion did not alter the clinical outcome and bacterial burden, although it moderately improved lung immunopathology. Treatment with a bacteriostatic antibiotic, azithromycin, alone significantly improved clinical outcome and bacterial clearance, but failed to reduce lung immunopathology. In comparison, treatment with a global inflammation inhibitor, dexamethasone, alone failed to alter clinical outcome, bacterial infection, and immunopathology, despite its moderate reducing effects on neutrophilic and cytokine responses. In contrast, combined treatment with both azithromycin and dexamethasone best improved clinical outcome, bacterial clearance, lung cellular and cytokine responses, and immunopathology. Our study suggests that marked improvement of clinical outcome and lung immunopathology caused by bacterial superinfection requires the control of both bacterial infection and aberrant host immune responses. Our findings hold implications in clinical management for influenza-associated bacterial superinfections.


Subject(s)
Immunity/immunology , Influenza A virus/physiology , Lung/immunology , Lung/pathology , Streptococcus pneumoniae/growth & development , Superinfection/microbiology , Superinfection/virology , Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Chemokine CXCL2/metabolism , Chemokines/metabolism , Disease Susceptibility/immunology , Disease Susceptibility/microbiology , Disease Susceptibility/pathology , Disease Susceptibility/virology , Female , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Immunity/drug effects , Immunotherapy , Influenza A virus/drug effects , Lung/microbiology , Lung/virology , Mice , Mice, Inbred C57BL , Neutralization Tests , Neutrophil Infiltration/drug effects , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/pathology , Pneumococcal Infections/drug therapy , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Streptococcus pneumoniae/drug effects , Superinfection/drug therapy , Superinfection/pathology , Treatment Outcome
11.
PLoS One ; 7(7): e41666, 2012.
Article in English | MEDLINE | ID: mdl-22844510

ABSTRACT

Tuberculosis (TB) vaccine-induced airway luminal T cells (ALT) have recently been shown to be critical to host defense against pulmonary TB. However, the mechanisms that maintain memory ALT remain poorly understood. In particular, whether respiratory mucosal exposure to environmental agents such as endotoxin may regulate the size of vaccine-induced ALT population is still unclear. Using a murine model of respiratory genetic TB vaccination and respiratory LPS exposure, we have addressed this issue in the current study. We have found that single or repeated LPS exposure increases the number of antigen-specific ALT which are capable of robust secondary responses to pulmonary mycobacterial challenge. To investigate the potential mechanisms by which LPS exposure modulates the ALT population, we have examined the role of ALT proliferation and peripheral T cell recruitment. We have found that LPS exposure-increased ALT is not dependent on increased ALT proliferation as respiratory LPS exposure does not significantly increase the rate of proliferation of ALT. But rather, we find it to be dependent upon the recruitment of peripheral T cells into the airway lumen as blockade of peripheral T cell supplies markedly reduces the initially increased ALT. Thus, our data suggest that environmental exposure to airborne agents such as endotoxin has a profound modulatory effect on TB vaccine-elicited T cells within the respiratory tract. Our study provides a new, M.tb antigen-independent mechanism by which the respiratory mucosal anti-TB memory T cells may be maintained.


Subject(s)
Endotoxins/toxicity , Respiratory System/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Animals , Antigens, Bacterial/immunology , Cell Proliferation/drug effects , Chemokines/metabolism , Female , Lipopolysaccharides/pharmacology , Mice , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory System/drug effects , Respiratory System/microbiology , Species Specificity , T-Lymphocytes/cytology , T-Lymphocytes/microbiology , Tuberculosis, Pulmonary/prevention & control
12.
Clin Immunol ; 144(1): 57-69, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22673491

ABSTRACT

Influenza epidemics and pandemics cause significant morbidity and mortality worldwide associated with severe immunopathology in the lung, and the mechanisms of such immunopathogenesis still remain poorly understood. While human studies help to understand influenza immunopathology, they provide only limited mechanistic information. On the other hand, recent studies using experimental animal models have significantly enhanced our understanding of the complex mechanisms involved in the immunopathogenesis during primary influenza or influenza-associated bacterial superinfection. This includes the involvement of acute inflammatory responses (macrophages, neutrophils, dendritic cells, toll-like receptors, cytokines, chemokines), CD4 and CD8 T cells, tissue remodeling processes, and contribution of bacterial superinfection. In particular, progress has been made in uncoupling the mechanisms that are involved in both anti-viral host defense and in immunopathogenesis from those that solely contribute to lung immunopathology. Uncoupling such events will facilitate the discovery of new intervention strategies to treat pulmonary immunopathology associated with influenza infection.


Subject(s)
Influenza, Human/immunology , Animals , Bacterial Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Inflammation/immunology , Superinfection/immunology
13.
Am J Pathol ; 179(6): 2963-76, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22001698

ABSTRACT

Lung immunopathology is the main cause of influenza-mediated morbidity and death, and much of its molecular mechanisms remain unclear. Whereas tumor necrosis factor-α (TNF-α) is traditionally considered a proinflammatory cytokine, its role in influenza immunopathology is unresolved. We have investigated this issue by using a model of acute H1N1 influenza infection established in wild-type and TNF-α-deficient mice and evaluated lung viral clearance, inflammatory responses, and immunopathology. Whereas TNF-α was up-regulated in the lung after influenza infection, it was not required for normal influenza viral clearance. However, TNF-α deficiency led not only to a greater extent of illness but also to heightened lung immunopathology and tissue remodeling. The severe lung immunopathology was associated with increased inflammatory cell infiltration, anti-influenza adaptive immune responses, and expression of cytokines such as monocyte chemoattractant protein-1 (MCP-1) and fibrotic growth factor, TGF-ß1. Thus, in vivo neutralization of MCP-1 markedly attenuated lung immunopathology and blunted TGF-ß1 production following influenza infection in these hosts. On the other hand, in vivo transgenic expression of MCP-1 worsened lung immunopathology following influenza infection in wild-type hosts. Thus, TNF-α is dispensable for influenza clearance; however, different from the traditional belief, this cytokine is critically required for negatively regulating the extent of lung immunopathology during acute influenza infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Viral/immunology , Tumor Necrosis Factor-alpha/physiology , Adaptive Immunity , Animals , Body Weight , Bronchoalveolar Lavage Fluid , Chemokine CCL2/deficiency , Chemokine CCL2/metabolism , Chemokines/metabolism , Cytokines/metabolism , Immunity, Cellular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/metabolism
14.
J Immunol ; 187(8): 4280-92, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21908731

ABSTRACT

Immunopathology is a major cause of influenza-associated morbidity and mortality worldwide. However, the role and regulatory mechanisms of CD4 T cells in severe lung immunopathology following acute influenza infection are poorly understood. In this paper, we report that the emergence of immunopathogenic CD4 T cells is under the control of a transmembrane immunoadaptor DAP12 pathway during influenza infection. We find that the mice lacking DAP12 have unaltered viral clearance but easily succumb to influenza infection as a result of uncontrolled immunopathology. Such immunopathology is associated with markedly increased CD4 T cells displaying markedly increased cytotoxicity and Fas ligand expression. Furthermore, the immunopathogenic property of these CD4 T cells is transferrable. Thus, depletion of CD4 T cells or abrogation of Fas/Fas ligand signaling pathway improves survival and immunopathology. We further find that DAP12 expressed by dendritic cells plays an important role in controlling the immunopathogenic CD4 T cells during influenza infection. Our findings identify a novel pathway that controls the level of immune-pathogenic CD4 T cells during acute influenza infection.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD4-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/pathology , Adoptive Transfer , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/virology , Respiratory Tract Infections/virology , Signal Transduction/immunology
15.
Am J Pathol ; 178(4): 1622-34, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21406169

ABSTRACT

The granuloma, a hallmark of host defense against pulmonary mycobacterial infection, has long been believed to be an active type 1 immune environment. However, the mechanisms regarding why granuloma fails to eliminate mycobacteria even in immune-competent hosts, have remained largely unclear. By using a model of pulmonary Mycobacterium bovis Bacillus Calmette-Guerin (BCG) infection, we have addressed this issue by comparing the immune responses within the airway luminal and granuloma compartments. We found that despite having a similar immune cellular profile to that in the airway lumen, the granuloma displayed severely suppressed type 1 immune cytokine but enhanced chemokine responses. Both antigen-presenting cells (APCs) and T cells in granuloma produced fewer type 1 immune molecules including tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), and nitric oxide. As a result, the granuloma APCs developed a reduced capacity to phagocytose mycobacteria and to induce T-cell proliferation. To examine the molecular mechanisms, we compared the levels of immune suppressive cytokine IL-10 in the airway lumen and granuloma and found that both granuloma APCs and T cells produced much more IL-10. Thus, IL-10 deficiency restored type 1 immune activation within the granuloma while having a minimal effect within the airway lumen. Hence, our study provides the first experimental evidence that, contrary to the conventional belief, the BCG-induced lung granuloma represents a symbiotic host-microbe microenvironment characterized by suppressed type 1 immune activation.


Subject(s)
Granuloma/microbiology , Interleukin-10/metabolism , Mycobacterium bovis/metabolism , Animals , Antigen-Presenting Cells/metabolism , BCG Vaccine/metabolism , CD11b Antigen/biosynthesis , CD11c Antigen/biosynthesis , Cell Proliferation , Female , Immune System , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Symbiosis , T-Lymphocytes/cytology , T-Lymphocytes/microbiology , Tumor Necrosis Factor-alpha/metabolism
16.
Blood ; 117(4): 1146-55, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21088134

ABSTRACT

We have recently reported that CD8(+) T-cell memory maintenance after immunization with recombinant human adenovirus type 5 (rHuAd5) is dependent upon persistent transgene expression beyond the peak of the response. In this report, we have further investigated the location and nature of the cell populations responsible for this sustained response. The draining lymph nodes were found to be important for primary expansion but not for memory maintenance, suggesting that antigen presentation through a nonlymphoid source was required. Using bone marrow chimeric mice, we determined that antigen presentation by nonhematopoietic antigen-presenting cells (APCs) was sufficient for maintenance of CD8(+) T-cell numbers. However, antigen presentation by this mechanism alone yielded a memory population that displayed alterations in phenotype, cytokine production and protective capacity, indicating that antigen presentation through both hematopoietic and nonhematopoietic APCs ultimately defines the memory CD8(+) T-cell response produced by rHuAd5. These results shed new light on the immunobiology of rHuAd5 vectors and provide evidence for a mechanism of CD8(+) T-cell expansion and memory maintenance that relies upon both hematopoietic and nonhematopoietic APCs.


Subject(s)
Adenoviruses, Human/immunology , CD8-Positive T-Lymphocytes/immunology , Immunization , Immunologic Memory/physiology , Viral Vaccines/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cells, Cultured , Female , Hematopoietic System/immunology , Humans , Immunization/methods , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Oncolytic Virotherapy/methods , Vaccines, Synthetic/immunology
17.
J Gene Med ; 12(8): 693-704, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20694950

ABSTRACT

BACKGROUND: Virus-vectored vaccine is a powerful activator of CD8 T cell-mediated immunity and is especially amenable to respiratory mucosal immunization, offering hopes for use in humans with diminished helper CD4 T cell function. However, whether virus-mediated mucosal immunization can produce immune protective CD8 T cells without the CD4 T cell help remains to be investigated. METHODS: We used a replication-deficient adenovirus vector expressing an Mycobacterium tuberculosis antigen Ag85A for intranasal vaccination and evaluated its effect on CD8 T cell activation and protection in mice depleted of CD4 T cells. RESULTS: Intranasal vaccination of CD4 T cell-depleted mice led to suboptimal generation of Ag-specific tetramer(+) or interferon (IFN)-gamma-producing CD8 T cells in the lung and spleen but this was observed mainly at the early time after vaccination. Reduced CD8 T cell priming was also accompanied by decreased CD8 T cell responses (CTL). Nevertheless, the ratio of Ag-specific CD8 T cells to IFN-gamma-producing CD8 T cells in CD4 T cell-depleted hosts remained comparable to that in CD4 T cell-competent hosts. Furthermore, the 'unhelped' CD8 T cells also displayed a similar immune phenotype as the 'helped' counterparts. The animals with 'unhelped' CD8 T cells were as well-protected from pulmonary M. tuberculosis challenge as those with 'helped' CD8 T cells in the absence of CD4 T cells. CONCLUSIONS: The data obtained in the present study suggest that the fully immune protective CD8 T cells can still be generated by respiratory mucosal viral-mediated immunization without CD4 T cells and that CD8 T cells, 'helped' or 'unhelped', can confer significant protection against pulmonary tuberculosis independent of CD4 T cells.


Subject(s)
Adenoviridae/genetics , CD8-Positive T-Lymphocytes/immunology , Respiratory Mucosa/immunology , Acyltransferases/immunology , Acyltransferases/metabolism , Adenoviridae/metabolism , Administration, Intranasal , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , CD8-Positive T-Lymphocytes/cytology , Cytotoxicity Tests, Immunologic , Female , Genetic Vectors , Immunization , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology
18.
J Immunol ; 184(4): 2048-56, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20083661

ABSTRACT

Influenza viral infection is well-known to predispose to subsequent bacterial superinfection in the lung but the mechanisms have remained poorly defined. We have established a murine model of heterologous infections by an H1N1 influenza virus and Staphylococcus aureus. We found that indeed prior influenza infection markedly increased the susceptibility of mice to secondary S. aureus superinfection. Severe sickness and heightened bacterial infection in flu and S. aureus dual-infected animals were associated with severe immunopathology in the lung. We further found that flu-experienced lungs had an impaired NK cell response in the airway to subsequent S. aureus bacterial infection. Thus, adoptive transfer of naive NK cells to the airway of prior flu-infected mice restored flu-impaired antibacterial host defense. We identified that TNF-alpha production of NK cells played an important role in NK cell-mediated antibacterial host defense as NK cells in flu-experienced lungs had reduced TNF-alpha expression and adoptive transfer of TNF-alpha-deficient NK cells to the airway of flu-infected mice failed to restore flu-impaired antibacterial host defense. Defected NK cell function was found to be an upstream mechanism of depressed antibacterial activities by alveolar macrophages as contrast to naive wild-type NK cells, the NK cells from flu-infected or TNF-alpha-deficient mice failed to enhance S. aureus phagocytosis by alveolar macrophages. Together, our study identifies the weakened NK cell response in the lung to be a novel critical mechanism for flu-mediated susceptibility to bacterial superinfection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Killer Cells, Natural/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/microbiology , Pneumonia, Bacterial/immunology , Pneumonia, Viral/immunology , Staphylococcal Infections/immunology , Superinfection/immunology , Animals , Disease Susceptibility/immunology , Disease Susceptibility/pathology , Female , Killer Cells, Natural/microbiology , Killer Cells, Natural/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/virology , Pneumonia, Viral/microbiology , Pneumonia, Viral/pathology , Staphylococcal Infections/pathology , Staphylococcal Infections/virology , Superinfection/microbiology , Superinfection/pathology , Superinfection/virology
19.
Am J Respir Crit Care Med ; 181(8): 862-72, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20019338

ABSTRACT

RATIONALE: The airway luminal memory CD8 T cells induced by respiratory mucosal immunization in a murine model have been found to be critical to antituberculosis immunity. However, the mechanisms of their maintenance on airway mucosal surface still remain poorly understood. OBJECTIVES: Using a model of adenovirus-based intranasal immunization we investigated the immune property and the mechanisms of maintenance of airway luminal CD8 T cells. METHODS: Immune properties of airway luminal Mycobacterium tuberculosis antigen-specific CD8 T cells were examined. Proliferation of airway luminal CD8 T cells was determined by in vivo T cell-labeling techniques. The role of peripheral T cell recruitment in maintaining airway luminal CD8 T cells was investigated by blocking lymphocyte trafficking from lymphoid and peripheral tissues. The requirement of M. tuberculosis antigens for in situ T cell proliferation was evaluated using a T cell transfer approach. An airway M. tuberculosis challenge model was used to study the relationship between CD8 T cell-mediated protection and peripheral T cell recruitment. MEASUREMENTS AND MAIN RESULTS: Intranasal immunization leads to elicitation of persisting M. tuberculosis antigen-specific CD8 T cells in the airway lumen, which display an activated effector memory phenotype different from those in peripheral tissues. Airway luminal T cells continuously proliferate in an antigen-dependent manner, and can be maintained even in the absence of peripheral T cell recruitment. The lungs equipped with such CD8 T cells are protected from airway M. tuberculosis challenge independent of both peripheral T cell supply and CD4 T cells. CONCLUSIONS: Vaccine-inducible airway luminal antituberculosis memory CD8 T cells are self-renewable in an antigen-dependent manner, and can be maintained independent of peripheral T cell supply.


Subject(s)
Bronchi/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Respiratory Mucosa/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Administration, Intranasal , Adoptive Transfer/methods , Animals , Antigens, Bacterial/drug effects , Antigens, Bacterial/immunology , Bronchi/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Immunization, Secondary , Immunologic Memory/drug effects , Lung/drug effects , Lung/immunology , Mice , Mice, Inbred BALB C , Respiratory Mucosa/drug effects , Tuberculosis, Pulmonary/immunology
20.
J Immunol ; 181(8): 5618-26, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832720

ABSTRACT

Protection by parenteral immunization with plasmid DNA vaccines against pulmonary tuberculosis (TB) is very modest. In this study, we have investigated the underlying mechanisms for the poor mucosal protective efficacy and the avenues and mechanisms to improve the efficacy of a single i.m. immunization with a monogenic plasmid DNA TB vaccine in a murine model. We show that i.m. DNA immunization fails to elicit accumulation of Ag-specific T cells in the airway lumen despite robust T cell responses in the spleen. Such systemically activated T cells cannot be rapidly mobilized into the airway lumen upon Mycobacterium tuberculosis exposure. However, airway deposition of low doses of soluble mycobacterial Ags in previously immunized mice effectively mobilizes the systemically activated T cells into the airway lumen. A fraction of such airway luminal T cells can persist in the airway lumen, undergo quick, robust expansion and activation and provide marked immune protection upon airway M. tuberculosis exposure. Airway mucosal deposition of soluble mycobacterial Ags was found to create a tissue microenvironment rich in proinflammatory molecules including chemokines and hence conducive to T cell recruitment. Thus, in vivo neutralization of MIP-1alpha or IFN-inducible protein-10 markedly inhibited the accumulation of Ag-specific T cells in the airway lumen. Our data suggest that immunoprotective efficacy on the mucosal surface by i.m. plasmid DNA immunization could be substantially improved by simple mucosal soluble Ag inoculation and restoration of mucosal luminal T cells. Our study holds implication for the future design of DNA vaccination strategies against intracellular infections.


Subject(s)
Antigens, Bacterial/pharmacology , Immunity, Mucosal/drug effects , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/pharmacology , Tuberculosis, Pulmonary/prevention & control , Vaccines, DNA/pharmacology , Administration, Intranasal , Animals , Antigens, Bacterial/immunology , Cell Movement/drug effects , Cell Movement/immunology , Chemokine CCL3/immunology , Chemokine CXCL10/immunology , Female , Humans , Immunity, Mucosal/immunology , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Plasmids/pharmacology , Respiratory System/immunology , T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Vaccination/methods , Vaccines, DNA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...