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










Publication year range
1.
J Immunol ; 194(5): 2199-207, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25646304

ABSTRACT

The development of therapeutic vaccines for treatment of established cancer has proven challenging. Cancer vaccines not only need to induce a robust tumor Ag-specific immune response but also need to overcome the tolerogenic and immunosuppressive microenvironments that exist within many solid cancers. ISCOMATRIX adjuvant (ISCOMATRIX) is able to induce both tumor Ag-specific cellular and Ab responses to protect mice against tumor challenge, but this is insufficient to result in regression of established solid tumors. In the current study, we have used B16-OVA melanoma, Panc-OVA pancreatic, and TRAMP-C1 prostate cancer mouse tumor models to test therapeutic efficacy of ISCOMATRIX vaccines combined with other immune modulators. The coadministration of an ISCOMATRIX vaccine with the TLR3 agonist, polyinosinic-polycytidylic acid, and TLR9 agonist, CpG, reduced tumor growth in all tumor models and the presence of ISCOMATRIX in the formulation was critical for the therapeutic efficacy of the vaccine. This vaccine combination induced a robust and multifunctional CD8(+) T cell response. Therapeutic protection required IFN-γ and CD8(+) T cells, whereas NK and CD4(+) T cells were found to be redundant. ISCOMATRIX vaccines combined with TLR3 and TLR9 agonists represent a promising cancer immunotherapy strategy.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/administration & dosage , Cholesterol/administration & dosage , Melanoma, Experimental/therapy , Pancreatic Neoplasms/therapy , Phospholipids/administration & dosage , Prostatic Neoplasms/therapy , Saponins/administration & dosage , Skin Neoplasms/therapy , Adjuvants, Immunologic/administration & dosage , Animals , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Drug Combinations , Humans , Immunotherapy/methods , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/mortality , Mice , Mice, Knockout , Neoplasm Transplantation , Oligodeoxyribonucleotides/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Poly I-C/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/mortality , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Survival Analysis , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Toll-Like Receptor 9/antagonists & inhibitors , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Tumor Burden/drug effects
2.
Expert Rev Vaccines ; 12(7): 733-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23885819

ABSTRACT

It is thought that the development of vaccines for the treatment of infectious diseases and cancer is likely to be achieved in the coming decades. This is partially due to a better understanding of the regulatory networks connecting innate with adaptive immune responses. The innate immune response is triggered by the recognition of conserved pathogen-associated molecular patterns by germ line-coded pattern recognition receptors. Several families of pattern recognition receptors have been characterized, including Toll-like receptors and nucleotide-binding domain receptors. The identification of their ligands has driven the development of novel adjuvants many of which have been tested in vaccine clinical trials. Here, the authors review recent preclinical data and clinical trial results supporting the view that combinations of adjuvants are the way forward in vaccine design. Multiadjuvanted vaccines can stimulate the broad and robust protective immune responses required to fight chronic infectious diseases and cancer.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Vaccination/methods , Vaccines/immunology , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Vaccines/administration & dosage
3.
J Immunol ; 190(9): 4585-94, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23536629

ABSTRACT

The transcription factor inhibitor of DNA binding (Id)2 modulates T cell fate decisions, but the molecular mechanism underpinning this regulation is unclear. In this study we show that loss of Id2 cripples effector differentiation and instead programs CD8(+) T cells to adopt a memory fate with increased Eomesodermin and Tcf7 expression. We demonstrate that Id2 restrains CD8(+) T cell memory differentiation by inhibiting E2A-mediated direct activation of Tcf7 and that Id2 expression level mirrors T cell memory recall capacity. As a result of the defective effector differentiation, Id2-deficient CD8(+) T cells fail to induce sufficient Tbx21 expression to generate short-lived effector CD8(+) T cells. Our findings reveal that the Id2/E2A axis orchestrates T cell differentiation through the induction or repression of downstream transcription factors essential for effector and memory T cell differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory/immunology , Inhibitor of Differentiation Protein 2/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Gene Expression/immunology , Hepatocyte Nuclear Factor 1-alpha , Immunologic Memory/genetics , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/immunology , Mice , Mice, Inbred C57BL , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/immunology , T Cell Transcription Factor 1/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism
4.
J Exp Med ; 209(11): 2049-64, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23045607

ABSTRACT

A strong humoral response to infection requires the collaboration of several hematopoietic cell types that communicate via antigen presentation, surface coreceptors and their ligands, and secreted factors. The proinflammatory cytokine IL-6 has been shown to promote the differentiation of activated CD4(+) T cells into T follicular helper cells (T(FH) cells) during an immune response. T(FH) cells collaborate with B cells in the formation of germinal centers (GCs) during T cell-dependent antibody responses, in part through secretion of critical cytokines such as IL-21. In this study, we demonstrate that loss of either IL-6 or IL-21 has marginal effects on the generation of T(FH) cells and on the formation of GCs during the response to acute viral infection. However, mice lacking both IL-6 and IL-21 were unable to generate a robust T(FH) cell-dependent immune response. We found that IL-6 production in follicular B cells in the draining lymph node was an important early event during the antiviral response and that B cell-derived IL-6 was necessary and sufficient to induce IL-21 from CD4(+) T cells in vitro and to support T(FH) cell development in vivo. Finally, the transcriptional activator Oct2 and its cofactor OBF-1 were identified as regulators of Il6 expression in B cells.


Subject(s)
B-Lymphocytes/immunology , Interleukin-6/immunology , Interleukins/immunology , Octamer Transcription Factor-2/immunology , T-Lymphocytes/immunology , Trans-Activators/immunology , Animals , Antibodies, Viral/immunology , B-Lymphocytes/metabolism , Blotting, Western , Flow Cytometry , Gene Expression Regulation/immunology , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/virology , Host-Pathogen Interactions/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/physiology , Interleukin-6/deficiency , Interleukin-6/genetics , Interleukins/genetics , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Octamer Transcription Factor-2/genetics , Octamer Transcription Factor-2/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
5.
EMBO J ; 30(13): 2690-704, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21587207

ABSTRACT

Dendritic cells (DCs) have critical roles in the induction of the adaptive immune response. The transcription factors Id2, Batf3 and Irf-8 are required for many aspects of murine DC differentiation including development of CD8α(+) and CD103(+) DCs. How they regulate DC subset specification is not completely understood. Using an Id2-GFP reporter system, we show that Id2 is broadly expressed in all cDC subsets with the highest expression in CD103(+) and CD8α(+) lineages. Notably, CD103(+) DCs were the only DC able to constitutively cross-present cell-associated antigens in vitro. Irf-8 deficiency affected loss of development of virtually all conventional DCs (cDCs) while Batf3 deficiency resulted in the development of Sirp-α(-) DCs that had impaired survival. Exposure to GM-CSF during differentiation induced expression of CD103 in Id2-GFP(+) DCs. It did not restore cross-presenting capacity to Batf3(-/-) or CD103(-)Sirp-α(-)DCs in vitro. Thus, Irf-8 and Batf3 regulate distinct stages in DC differentiation during the development of cDCs. Genetic mapping DC subset differentiation using Id2-GFP may have broad implications in understanding the interplay of DC subsets during protective and pathological immune responses.


Subject(s)
Antigens, CD/metabolism , CD8 Antigens/metabolism , Cell Lineage/genetics , Dendritic Cells/physiology , Inhibitor of Differentiation Protein 2/genetics , Integrin alpha Chains/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Dendritic Cells/metabolism , Gene Expression/physiology , Genes, cdc/physiology , Inhibitor of Differentiation Protein 2/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological
6.
J Immunol ; 186(7): 4059-66, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339359

ABSTRACT

Upon Ag encounter, naive T cells undergo extensive Ag-driven proliferation and can differentiate into effector cells. Up to 95% of these cells die leaving a small residual population of T cells that provide protective memory. In this study, we investigated the contribution of the BH3-only family protein Bid in the shutdown of T cell responses after acute and persistent infection. Influenza virus pathogenicity has been proposed to be mediated by a peptide encoded in the basic polymerase (PB1-RF2) acting through Bid. In our experiments, we found that after acute infection with influenza virus, mice lacking Bid had normal expansion and contraction of Ag-specific CD8(+) T cells. However, in chronic γ-herpesvirus infection, Bid-deficient virus-specific CD8(+) T cells expanded normally but failed to contract fully and were maintained at ∼2-fold higher levels. Previously, we have demonstrated that Bim plays a prominent role in T cell shutdown in persistent infection by cooperating with the death receptor Fas, which regulates apoptosis in response to repeated TCR signaling. Bid lies at the nexus of these two signaling pathways, thus we reasoned that Bid and Bim might cooperate in regulation of T cell shutdown in persistent infection. In this study, we observed that the combined loss of Bid and Bim synergistically enhanced the persistence of CD8(+) T cells during γ-herpesvirus infection. Thus, these data uncover a role for Bid in coordinating apoptotic signaling pathways to ensure appropriate shutdown of T cell immune responses in the setting of persistent Ag exposure.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/immunology , BH3 Interacting Domain Death Agonist Protein/physiology , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Lymphocyte Cooperation/immunology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Animals , Bcl-2-Like Protein 11 , Cell Communication/immunology , Cell Death/immunology , Herpesviridae Infections/metabolism , Influenza A Virus, H3N2 Subtype/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Rhadinovirus/immunology , T-Lymphocyte Subsets/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Viral Load/immunology
7.
J Immunol ; 185(6): 3669-76, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20720208

ABSTRACT

A critical factor influencing the ability of the host to mount a robust immune response against a virus depends on the rapid recruitment of dendritic cells (DCs) presenting Ags. From the outset, this step sets the tempo for subsequent activation of virus-specific T cells. Despite this, how induction of the immune response might be modified by pathogens with the capacity to establish persistence is unclear. In this study, we have characterized the in vivo influence of murine gamma-herpesvirus K3-mediated interference with MHC class I in DCs that drive the initial adaptive immune response. We observed that gamma-herpesvirus could interfere with the very earliest phase of Ag presentation through K3 by directly targeting migratory and lymph node-resident DCs. These results show that a pathogen with the capacity to interfere with early Ag presentation can establish suboptimal conditions for rapid induction of the adaptive immune response and thus favor establishment of viral persistence.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Herpesviridae Infections/immunology , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Animals , Chronic Disease , Cross-Priming/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Herpesviridae Infections/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rhadinovirus/pathogenicity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Tumor Virus Infections/metabolism , Viral Interference/immunology , Viral Proteins/antagonists & inhibitors , Viral Proteins/biosynthesis
8.
Methods Mol Biol ; 595: 299-318, 2010.
Article in English | MEDLINE | ID: mdl-19941121

ABSTRACT

Respiratory viral infections are a major cause of morbidity and mortality. Protection of the respiratory tract from pathogen infections, such as influenza virus, requires the orchestrated activation and trafficking of pulmonary dendritic cells (DCs) from the lung to the lymph node (LN) in order to ensure optimized T-cell responses. Gaining a better understanding of the cellular and molecular processes that protect the lung during infection is essential for future advances in vaccine strategies and treatments. Influenza viral infection in mice offers a very well-defined immunological system in which the underlying parameters regulating the generation of protective immunity can be elucidated. In this chapter, we review methods for quantitative analysis of DC and T-cell responses in a murine model infection of influenza. Antigen-specific tracking and quantitation of viral immune responses have been greatly facilitated by the advent of MHC tetramers and intracellular cytokine analysis, together with gentle isolation procedures for dendritic cells allowing detection of viral and endogenous antigens.


Subject(s)
Lung/immunology , Lung/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Animals , Antigen Presentation/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Disease Models, Animal , Lymphocyte Activation/immunology , Mice , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/virology , Respiratory Tract Infections , T-Lymphocytes/cytology , T-Lymphocytes/immunology
9.
Immunity ; 30(2): 218-27, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19200758

ABSTRACT

CD40L (CD154) on CD4(+) T cells has been shown to license dendritic cells (DCs) via CD40 to prime cytotoxic T lymphocyte (CTL) responses. We found that the converse (CD40L on DCs) was also important. Anti-CD40L treatment decreased endogenous CTL responses to both ovalbumin and influenza infection even in the absence of CD4(+) T cells. DCs expressed CD40L upon stimulation with agonists to Toll-like receptor 3 (TLR3) and TLR9. Moreover, influenza infection, which stimulates CTLs without help, upregulated CD40L on DCs, but herpes simplex infection, which elicits CTLs through help, did not. CD40L-deficient (Cd40lg(-/-)) DCs are suboptimal both in vivo in bone marrow chimera experiments and in vitro in mixed lymphocyte reactions. In contrast, Cd40lg(-/-) CD8(+) T cells killed as effectively as wild-type cells. Thus, CD40L upregulation on DCs promoted optimal priming of CD8(+) T cells without CD4(+) T cells, providing a mechanism by which pathogens may elicit helper-independent CTL immunity.


Subject(s)
CD40 Ligand/immunology , Dendritic Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptors/metabolism , Animals , Antibodies/immunology , CD40 Antigens/immunology , CD40 Ligand/deficiency , CD40 Ligand/genetics , CD40 Ligand/metabolism , Dendritic Cells/metabolism , Ligands , Mice , Mice, Knockout , T-Lymphocytes, Cytotoxic/virology , Toll-Like Receptors/agonists , Up-Regulation/drug effects , Up-Regulation/immunology
10.
Handb Exp Pharmacol ; (188): 51-77, 2009.
Article in English | MEDLINE | ID: mdl-19031021

ABSTRACT

Antigen presenting cells (APCs) are recognized as key initiators of adaptive immunity, particularly to pathogens, by eliciting a rapid and potent immune attack on infected cells. Amongst APCs, dendritic cells (DCs) are specially equipped to initiate and regulate immune responses in a manner that depends on signals they receive from microbes and their cellular environment. To achieve this, they are equipped with highly efficient mechanisms that allow them to detect pathogens, to capture, process and present antigens, and to activate and guide the differentiation of T cells into effector and memory cells. DCs can no longer be considered as a homogeneous cell type performing a single function, but are heterogeneous both in phenotype, function and dependence on inflammatory stimuli for their formation and responsiveness. Recent studies of DC subtypes have highlighted the contrasting roles of different professional APCs in activating divergent arms of the immune response towards pathogens. In this review, we discuss the progress that has been made in dissecting the attributes of different DC subsets that migrate into, or reside permanently, within lymphoid tissues and their putative roles in the induction of the anti-viral immune response.


Subject(s)
Antigen Presentation , Antigens, Viral/immunology , Dendritic Cells/immunology , Virus Diseases/immunology , Animals , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology
11.
Nat Immunol ; 9(11): 1244-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18849989

ABSTRACT

The importance of conventional dendritic cells (cDCs) in the processing and presentation of antigen is well established, but the contribution of plasmacytoid dendritic cells (pDCs) to these processes, and hence to T cell immunity, remains unclear. Here we showed that unlike cDCs, pDCs continued to synthesize major histocompatibility complex (MHC) class II molecules and the MHC class II ubiquitin ligase MARCH1 long after activation. Sustained MHC class II-peptide complex formation, ubiquitination and turnover rendered pDCs inefficient in the presentation of exogenous antigens but enabled pDCs to continuously present endogenous viral antigens in their activated state. As the antigen-presenting abilities of cDCs and pDCs are fundamentally distinct, these two cell types may activate largely nonoverlapping repertoires of CD4(+) T cells.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Histocompatibility Antigens Class II/metabolism , Ubiquitination , Animals , Antigens, Viral/immunology , CD11 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Histocompatibility Antigens Class II/biosynthesis , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , Mice , Mice, Inbred Strains , Mice, Knockout , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
12.
Immunol Cell Biol ; 86(4): 333-42, 2008.
Article in English | MEDLINE | ID: mdl-18347609

ABSTRACT

Protective immunity against viral pathogens depends on the generation and maintenance of a small population of memory CD8(+) T cells. Successful memory cell generation begins with early interactions between naïve T cell and dendritic cells (DCs) within the inflammatory milieu of the secondary lymphoid tissues. Recent insights into the role of different populations of DCs, and kinetics of antigen presentation, during viral infections have helped to understand how DCs can shape the immune response. Here, we review the recent progress that has been made towards defining how specific DC subsets drive effector CD8(+) T-cell expansion and differentiation into memory cells. Further, we endeavour to examine how the molecular signals imparted by DCs coordinate to generate protective CD8(+) T-cell immunity.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Virus Diseases/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Inflammation/virology
13.
PLoS One ; 3(2): e1691, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18301768

ABSTRACT

Dendritic cells (DC) are a heterogeneous cell population that bridge the innate and adaptive immune systems. CD8alpha DC play a prominent, and sometimes exclusive, role in driving amplification of CD8(+) T cells during a viral infection. Whether this reliance on a single subset of DC also applies for CD4(+) T cell activation is unknown. We used a direct ex vivo antigen presentation assay to probe the capacity of flow cytometrically purified DC populations to drive amplification of CD4(+) and CD8(+) T cells following infection with influenza virus by different routes. This study examined the contributions of non-CD8alpha DC populations in the amplification of CD8(+) and CD4(+) T cells in cutaneous and systemic influenza viral infections. We confirmed that in vivo, effective immune responses for CD8(+) T cells are dominated by presentation of antigen by CD8alpha DC but can involve non-CD8alpha DC. In contrast, CD4(+) T cell responses relied more heavily on the contributions of dermal DC migrating from peripheral lymphoid tissues following cutaneous infection, and CD4 DC in the spleen after systemic infection. CD4(+) T cell priming by DC subsets that is dependent upon the route of administration raises the possibility that vaccination approaches could be tailored to prime helper T cell immunity.


Subject(s)
Antigen Presentation , CD4-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Immunity , Mice , Orthomyxoviridae , Skin/pathology , Skin/virology , Spleen/pathology , Spleen/virology
14.
Proc Natl Acad Sci U S A ; 104(45): 17753-8, 2007 Nov 06.
Article in English | MEDLINE | ID: mdl-17978177

ABSTRACT

When dendritic cells (DCs) encounter signals associated with infection or inflammation, they become activated and undergo maturation. Mature DCs are very efficient at presenting antigens captured in association with their activating signal but fail to present subsequently encountered antigens, at least in vitro. Such impairment of MHC class II (MHC II) antigen presentation has generally been thought to be a consequence of down-regulation of endocytosis, so it might be expected that antigens synthesized by the DCs themselves (for instance, viral antigens) would still be presented by mature DCs. Here, we show that DCs matured in vivo could still capture and process soluble antigens, but were unable to present peptides derived from these antigens. Furthermore, presentation of viral antigens synthesized by the DCs themselves was also severely impaired. Indeed, i.v. injection of pathogen mimics, which caused systemic DC activation in vivo, impaired the induction of CD4 T cell responses against subsequently encountered protein antigens. This immunosuppressed state could be reversed by adoptive transfer of DCs loaded exogenously with antigens, demonstrating that impairment of CD4 T cell responses was due to lack of antigen presentation rather than to overt suppression of T cell activation. The biochemical mechanism underlying this phenomenon was the down-regulation of MHC II-peptide complex formation that accompanied DC maturation. These observations have important implications for the design of prophylactic and therapeutic DC vaccines and contribute to the understanding of the mechanisms causing immunosuppression during systemic blood infections.


Subject(s)
Antigens, Viral/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Viral Vaccines/immunology , Animals , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/virology , Flow Cytometry , Mice , Muramidase/immunology , Ovalbumin/immunology
15.
J Immunol ; 179(11): 7506-13, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18025195

ABSTRACT

The activation and maintenance of Ag-specific CD8(+) T cells is central to the long-term control of persistent infections. These killer T cells act to continuously scan and remove reservoirs of pathogen that have eluded the acute immune response. Acutely cleared viral infections depend almost exclusively on dendritic cells (DC) to present Ags to, and to activate, the CD8(+) T cell response. Paradoxically, persistent pathogens often infect professional APCs such as DC, in addition to infecting a broad range of nonprofessional APC, raising the possibility that many cell types could present viral Ags and activate T cells. We addressed whether in persistent viral infection with murine gammaherpesviruses, DC or non-DC, such as B cells and macrophages, were required to maintain the continued activation of Ag-specific CD8(+) T cells. We found that presentation of the surrogate Ag, OVA, expressed under a lytic promoter to CD8(+) T cells during persistent infection was largely restricted to DC, with little contribution from other lymphoid resident cells, such as B cells. This is despite the fact that B cells harbor a very large reservoir of latent virus. Our results support that, during persistent viral infection, continual presentation of lytic Ags by DC leads to T cell activation critical for maintaining CD8(+) T cells capable of limiting persistent viral infection.


Subject(s)
Antigen Presentation/immunology , Antigens, Viral/immunology , Dendritic Cells/immunology , Gammaherpesvirinae/immunology , Herpesviridae Infections/immunology , Animals , CD11c Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Dendritic Cells/virology , Immunologic Memory/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Eur J Immunol ; 36(12): 3186-97, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17109468

ABSTRACT

CD4(+) T cells play a major role in containing herpesvirus infections. However, their cellular targets remain poorly defined. In vitro CD4(+) T cells have been reported to kill B cells that harbor a latent gammaherpesvirus. We used the B cell-tropic murine gammaherpesvirus-68 (MHV-68) to test whether this also occurred in vivo. MHV-68 that expressed cytoplasmic ovalbumin (OVA) in tandem with its episome maintenance protein, ORF73, stimulated CD8(+) T cells specific for the H2-K(b)-restricted OVA epitope SIINFEKL and was rapidly eliminated from C57BL/6 (H2(b)) mice. However, the same virus failed to stimulate CD4(+) T cells specific for the I-A(d)/I-A(b)-restricted OVA(323-339) epitope. We overcame any barrier to the MHC class II-restricted presentation of an endogenous epitope by substituting OVA(323-339) for the CLIP peptide of the invariant chain (ORF73-IRES-Ii-OVA), again expressed in tandem with ORF73. This virus presented OVA(323-339) but showed little or no latency deficit in either BALB/c (H2(d)) or C57BL/6 mice. Latent antigen-specific CD4(+) T cells therefore either failed to recognize key virus-infected cell populations in vivo or lacked the effector functions required to control them.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Herpesviridae Infections/immunology , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Virus Latency/immunology , Animals , Cell Line , Cells, Cultured , Mice , Mice, Inbred BALB C , Models, Biological , NIH 3T3 Cells
18.
Eur J Immunol ; 36(2): 327-35, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16402408

ABSTRACT

While naive CD8(+) T cells have been shown to require bone marrow-derived dendritic cells (DC) to initiate immunity, such a requirement for memory CD8(+) T cells has had limited assessment. By generating bone marrow chimeras that express the appropriate antigen-presenting molecules on either radiation-sensitive bone marrow-derived or radiation-resistant non-bone marrow-derived compartments, we showed that both primary and secondary immune responses to influenza virus infection of the lung were initiated in the draining LN. This required cells of bone marrow origin, most likely DC, for optimal expansion within the secondary lymphoid compartment. This was similarly the case with HSV-1 infection of the skin. As Langerhans cells are radioresistant, unlike other DC populations, these studies also demonstrate that the radiosensitive DC responsible for secondary expansion of HSV-specific memory are not Langerhans cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/immunology , Immunologic Memory/immunology , Lung Diseases/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Simplexvirus/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Female , Herpes Simplex/genetics , Immunologic Memory/genetics , Immunologic Memory/radiation effects , Langerhans Cells/immunology , Lung Diseases/genetics , Lymph Nodes/immunology , Male , Mice , Mice, Knockout , Orthomyxoviridae/genetics , Orthomyxoviridae Infections/genetics , Transplantation Chimera , Whole-Body Irradiation
19.
Nat Immunol ; 7(2): 165-72, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415871

ABSTRACT

The mechanisms responsible for the immunosuppression associated with sepsis or some chronic blood infections remain poorly understood. Here we show that infection with a malaria parasite (Plasmodium berghei) or simple systemic exposure to bacterial or viral Toll-like receptor ligands inhibited cross-priming. Reduced cross-priming was a consequence of downregulation of cross-presentation by activated dendritic cells due to systemic activation that did not otherwise globally inhibit T cell proliferation. Although activated dendritic cells retained their capacity to present viral antigens via the endogenous major histocompatibility complex class I processing pathway, antiviral responses were greatly impaired in mice exposed to Toll-like receptor ligands. This is consistent with a key function for cross-presentation in antiviral immunity and helps explain the immunosuppressive effects of systemic infection. Moreover, inhibition of cross-presentation was overcome by injection of dendritic cells bearing antigen, which provides a new strategy for generating immunity during immunosuppressive blood infections.


Subject(s)
Dendritic Cells/immunology , Malaria/immunology , Toll-Like Receptors/metabolism , Animals , Antigen Presentation , Antigens, Viral , CpG Islands/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Immune Tolerance , In Vitro Techniques , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Plasmodium berghei , T-Lymphocytes, Cytotoxic/immunology
20.
Lancet ; 363(9424): 1860-7, 2004 Jun 05.
Article in English | MEDLINE | ID: mdl-15183624

ABSTRACT

BACKGROUND: HIV infection increases the risk of malaria infection in pregnant women. Antibodies to variant surface antigens (VSA) on infected erythrocytes might protect against malaria in pregnancy. We postulated that HIV-induced impairment of humoral immunity to VSA mediates the increased susceptibility to malaria. METHODS: We compared serum concentrations of antibodies to VSA by flow cytometry or agglutination, and to merozoite proteins AMA-1 and MSP119 by ELISA, in 298 pregnant Malawian women, and related the findings to malaria and HIV infection, CD4-positive T-cell count, and HIV-1 viral load. FINDINGS: Concentrations of IgG to placental type VSA were lower in HIV-infected women than in HIV-uninfected women (median 8 units [IQR 4-23] vs 20 [12-30]; p<0.0001), among women with malaria (p=0.009) and those without malaria (p=0.0062). The impairment was greatest in first pregnancy. Agglutinating antibodies to placental VSA were present in a lower proportion of HIV-infected than HIV-uninfected women (58 [35.1%] of 165 vs 50 [53.8%] of 93, p<0.001). The degree of antibody binding by flow cytometry was correlated with CD4-positive T-cell count (r=0.16, p=0.019) and inversely with HIV-1 viral load (r=-0.16, p=0.030). Concentrations of antibodies to AMA-1 were lower in HIV infection (p<0.0001) but were not correlated with CD4-positive T-cell count or viral load. Responses to MSP119 were little affected by HIV infection. In multivariate analyses, HIV was negatively associated with amount of antibody to both VSA and AMA-1 (p<0.001 for each) but not MSP119. INTERPRETATION: HIV infection impairs antimalarial immunity, especially responses to placental type VSA. The impairment is greatest in the most immunosuppressed women and could explain the increased susceptibility to malaria seen in pregnant women with HIV infection.


Subject(s)
Antigens, Protozoan/immunology , HIV Infections/immunology , HIV-1 , Malaria, Falciparum/immunology , Pregnancy Complications, Infectious/immunology , Adult , Animals , Antibodies, Protozoan/blood , Antigens, Surface/immunology , CD4 Lymphocyte Count , Chondroitin Sulfates/immunology , Enzyme-Linked Immunosorbent Assay , Erythrocyte Membrane/immunology , Erythrocytes/parasitology , Female , Gravidity , HIV Infections/complications , Humans , Immunoglobulin G/blood , Malaria, Falciparum/complications , Malaria, Falciparum/parasitology , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Pregnancy , Pregnancy Complications, Parasitic/immunology , Protein Subunits/immunology , Protozoan Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...