Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Res ; 73(22): 6597-608, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24048821

ABSTRACT

Antitumor immunity is strongly influenced by the balance of tumor antigen-specific effector T cells (Teff) and regulatory T cells (Treg). However, the impact that vaccine adjuvants have in regulating the balance of antigen-specific T-cell populations is not well understood. We found that antigen-specific Tregs were induced following subcutaneous vaccination with either OVA or melanoma-derived peptides, with a restricted expansion of Teffs. Addition of the adjuvants CpG-ODN or Poly(I:C) preferentially amplified Teffs over Tregs, dramatically increasing the antigen-specific Teff:Treg ratios and inducing polyfunctional effector cells. In contrast, two other adjuvants, imiquimod and Quil A saponin, favored an expansion of antigen-specific Tregs and failed to increase Teff:Treg ratios. Following therapeutic vaccination of tumor-bearing mice, high ratios of tumor-specific Teffs:Tregs in draining lymph nodes were associated with enhanced CD8(+) T-cell infiltration at the tumor site and a durable rejection of tumors. Vaccine formulations of peptide+CpG-ODN or Poly(I:C) induced selective production of proinflammatory type I cytokines early after vaccination. This environment promoted CD8(+) and CD4(+) Teff expansion over that of antigen-specific Tregs, tipping the Teff to Treg balance to favor effector cells. Our findings advance understanding of the influence of different adjuvants on T-cell populations, facilitating the rational design of more effective cancer vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Cellular/drug effects , Neoplasms/immunology , T-Cell Antigen Receptor Specificity/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Adjuvants, Immunologic/administration & dosage , Animals , Antigens/immunology , Cancer Vaccines/administration & dosage , Female , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/therapy , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Treatment Outcome , Tumor Cells, Cultured
2.
PLoS One ; 7(4): e35005, 2012.
Article in English | MEDLINE | ID: mdl-22506061

ABSTRACT

The expression of melanoma-associated antigens (MAA) being limited to normal melanocytes and melanomas, MAAs are ideal targets for immunotherapy and melanoma vaccines. As MAAs are derived from self, immune responses to these may be limited by thymic tolerance. The extent to which self-tolerance prevents efficient immune responses to MAAs remains unknown. The autoimmune regulator (AIRE) controls the expression of tissue-specific self-antigens in thymic epithelial cells (TECs). The level of antigens expressed in the TECs determines the fate of auto-reactive thymocytes. Deficiency in AIRE leads in both humans (APECED patients) and mice to enlarged autoreactive immune repertoires. Here we show increased IgG levels to melanoma cells in APECED patients correlating with autoimmune skin features. Similarly, the enlarged T cell repertoire in AIRE(-/-) mice enables them to mount anti-MAA and anti-melanoma responses as shown by increased anti-melanoma antibodies, and enhanced CD4(+) and MAA-specific CD8(+) T cell responses after melanoma challenge. We show that thymic expression of gp100 is under the control of AIRE, leading to increased gp100-specific CD8(+) T cell frequencies in AIRE(-/-) mice. TRP-2 (tyrosinase-related protein), on the other hand, is absent from TECs and consequently TRP-2 specific CD8(+) T cells were found in both AIRE(-/-) and AIRE(+/+) mice. This study emphasizes the importance of investigating thymic expression of self-antigens prior to their inclusion in vaccination and immunotherapy strategies.


Subject(s)
Autoantigens/immunology , Immune Tolerance/immunology , Melanoma, Experimental/immunology , Melanoma/immunology , Thymocytes/immunology , gp100 Melanoma Antigen/immunology , Animals , Antibodies, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Epithelial Cells/immunology , Female , Humans , Immunoglobulin G/immunology , Immunologic Factors/immunology , Intramolecular Oxidoreductases/immunology , Melanoma/therapy , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyendocrinopathies, Autoimmune/blood , Polyendocrinopathies, Autoimmune/immunology , Self Tolerance/immunology
3.
Vaccine ; 30(2): 388-97, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22079266

ABSTRACT

Protein and peptide-based vaccines provide rigorously formulated antigens. However, these purified products are only weakly immunogenic by themselves and therefore require the addition of immunostimulatory components or adjuvants in the vaccine formulation. Various compounds derived from pathogens, minerals or plants, possess pro-inflammatory properties which allow them to act as adjuvants and contribute to the induction of an effective immune response. The results presented here demonstrate the adjuvant properties of novel saponins derived from the Spanish saffron Crocus sativus. In vivo immunization studies and tumor protection experiments unambiguously establish the value of saffron saponins as candidate adjuvants. These saponins were indeed able to increase both humoral and cellular immune responses to protein-based vaccines, ultimately providing a significant degree of protection against tumor challenge when administered in combination with a tumor antigen. This preclinical study provides an in depth immunological characterization of a new saponin as a vaccine adjuvant, and encourages its further development for use in vaccine formulations.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/isolation & purification , Crocus/chemistry , Saponins/administration & dosage , Saponins/isolation & purification , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Immunity, Cellular , Immunity, Humoral , Male , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/prevention & control
4.
Eur J Immunol ; 41(8): 2217-28, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21538347

ABSTRACT

In the last two decades, anti-cancer vaccines have yielded disappointing clinical results despite the fact that high numbers of self/tumor-specific T cells can be elicited in immunized patients. Understanding the reasons behind this lack of efficacy is critical in order to design better treatment regimes. Recombinant lentivectors (rLVs) have been successfully used to induce antigen-specific T cells to foreign or mutated tumor antigens. Here, we show that rLV expressing a murine nonmutated self/tumor antigen efficiently primes large numbers of self/tumor-specific CD8(+) T cells. In spite of the large number of tumor-specific T cells, however, no anti-tumor activity could be measured in a therapeutic setting, in mice vaccinated with rLV. Accumulating evidence shows that, in the presence of malignancies, inhibition of T-cell activity may predominate overstimulation. Analysis of tumor-infiltrating lymphocytes revealed that specific anti-tumor CD8(+) T cells fail to produce cytokines and express high levels of inhibitory receptors such as programmed death (PD)-1. Association of active immunization with chemotherapy or antibodies that block inhibitory pathways often leads to better anti-tumor effects. We show here that combining rLV vaccination with either cyclophosphamide or PD-1 and PD-L1 blocking antibodies enhances rLV vaccination efficacy and improves anti-tumor immunity.


Subject(s)
Antibodies, Blocking/pharmacology , Antineoplastic Agents/pharmacology , Immunization/methods , Lentivirus/immunology , Melanoma, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Blocking/immunology , Antigens, Differentiation/immunology , B7-1 Antigen/immunology , B7-H1 Antigen , Cell Line, Tumor , Combined Modality Therapy , Cyclophosphamide/pharmacology , Dose-Response Relationship, Drug , Epitopes/immunology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Intramolecular Oxidoreductases/immunology , Lentivirus/genetics , Lymphocyte Count , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Peptides/immunology , Programmed Cell Death 1 Receptor , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
5.
J Immunol ; 186(7): 3874-81, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21357256

ABSTRACT

CMV establishes a lifelong persistent infection, and viral immune-modulating strategies are important in facilitating this. A particularly diverse CD8 T cell response develops as a result of this host-virus détente, with the CMV-specific memory T cell pool displaying unique functions and phenotypes. To gain insight into the factors that regulate CMV-specific CD8 T cell responses, we examined the influence of the B7-CD28 costimulatory pathway on magnitude, kinetics, and phenotype. Initial expansion of mouse CMV-specific CD8 T cells that establish stable memory pools was severely lower in mice lacking B7-CD28 signaling, and the resulting memory levels also remained reduced during persistent/latent infection. In contrast, expansion of CD8 T cells that undergo memory inflation during chronic infection was less affected in the absence of B7-CD28 costimulatory signals, eventually reaching the levels seen in wild-type mice at later times. Regardless of their differential requirements for B7-CD28 signals, both stable and inflationary memory T cell populations showed normal cytotoxic capacity. These results reveal that B7-CD28 costimulation differentially regulates the magnitude and kinetics of the multifaceted CD8 T cell response that develops during CMV infection.


Subject(s)
B7-1 Antigen/physiology , B7-2 Antigen/physiology , CD28 Antigens/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Immunologic Memory , Muromegalovirus/immunology , Adaptive Immunity/genetics , Animals , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , CD28 Antigens/genetics , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Immunologic Memory/genetics , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology
6.
Expert Opin Ther Targets ; 15(1): 91-101, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21142803

ABSTRACT

IMPORTANCE OF THE FIELD: Promising immunotherapeutic agents targeting co-stimulatory pathways are currently being tested in clinical trials. One player in this array of regulatory pathways is the LAG-3/MHC class II axis. The lymphocyte activation gene-3 (LAG-3) is a negative co-stimulatory receptor that modulates T cell homeostasis, proliferation and activation. A recombinant soluble dimeric form of LAG-3 (sLAG-3-Ig, IMP321) shows adjuvant properties and enhances immunogenicity of tumor vaccines. Recent clinical trials produced encouraging results, especially when the human dimeric soluble form of LAG-3 (hLAG-3-Ig) was used in combination with chemotherapy. AREAS COVERED IN THIS REVIEW: The biological relevance of LAG-3 in vivo. Pre-clinical data demonstrating adjuvant properties, as well as the improvement of tumor immunity by sLAG-3-Ig. Recent advances in the clinical development of the therapeutic reagent IMP321, hLAG-3-Ig, for cancer treatment. WHAT THE READER WILL GAIN: This review summarizes preclinical and clinical data on the biological functions of LAG-3. TAKE HOME MESSAGE: The LAG-3 inhibitory pathway is attracting attention, in the light of recent studies demonstrating its role in T cell unresponsiveness, and Treg function after chronic antigen stimulation. As a soluble recombinant dimer, the sLAG-3-Ig protein acts as an adjuvant for therapeutic induction of T cell responses, and may be beneficial to cancer patients when used in combination therapies.


Subject(s)
Antigens, CD/immunology , Cancer Vaccines/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, CD/administration & dosage , Humans , Lymphocyte Activation/immunology , Neoplasms/immunology , Solubility , T-Lymphocytes, Regulatory/immunology , Lymphocyte Activation Gene 3 Protein
7.
mBio ; 1(4)2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20844609

ABSTRACT

Efficient vaccination against infectious agents and tumors depends on specific antigen targeting to dendritic cells (DCs). We report here that biosafe coronavirus-based vaccine vectors facilitate delivery of multiple antigens and immunostimulatory cytokines to professional antigen-presenting cells in vitro and in vivo. Vaccine vectors based on heavily attenuated murine coronavirus genomes were generated to express epitopes from the lymphocytic choriomeningitis virus glycoprotein, or human Melan-A, in combination with the immunostimulatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF). These vectors selectively targeted DCs in vitro and in vivo resulting in vector-mediated antigen expression and efficient maturation of DCs. Single application of only low vector doses elicited strong and long-lasting cytotoxic T-cell responses, providing protective antiviral and antitumor immunity. Furthermore, human DCs transduced with Melan-A-recombinant human coronavirus 229E efficiently activated tumor-specific CD8(+) T cells. Taken together, this novel vaccine platform is well suited to deliver antigens and immunostimulatory cytokines to DCs and to initiate and maintain protective immunity.


Subject(s)
Antigens/administration & dosage , Cancer Vaccines/immunology , Coronavirus/genetics , Dendritic Cells/immunology , Genetic Vectors/genetics , Neoplasms/prevention & control , Viral Vaccines/immunology , Virus Diseases/prevention & control , Animals , Antigens/genetics , Antigens/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line , Cells, Cultured , Coronavirus/immunology , Dendritic Cells/virology , Genetic Vectors/immunology , Humans , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Species Specificity , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Diseases/immunology
8.
Infect Immun ; 76(5): 2164-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18332207

ABSTRACT

The extracellular adhesion protein (Eap) secreted by the major human pathogen Staphylococcus aureus is known to have several effects on human immunity. We have recently added to knowledge of these roles by demonstrating that Eap enhances interactions between major histocompatibility complex molecules and human leukocytes. Several studies have indicated that Eap can induce cytokine production by human peripheral blood mononuclear cells (PBMCs). To date, there has been no rigorous attempt to identify the breadth of cytokines produced by Eap stimulation or to identify the cell subsets that respond. Here, we demonstrate that Eap induces the secretion of the proinflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-alpha) by CD14(+) leukocytes (monocytes and macrophages) within direct ex vivo PBMC populations (note that granulocytes are also CD14(+) but are largely depleted from PBMC preparations). Anti-intercellular adhesion molecule 1 (CD54) antibodies inhibited this induction and implicated a role for this known Eap binding protein in cellular activation. IL-6 and TNF-alpha secretion by murine cells exposed to Eap was also observed. The activation of CD14(+) cells by Eap suggests that it could play a significant role in both septic shock and fever, two of the major pathological features of S. aureus infections.


Subject(s)
Bacterial Proteins/immunology , Interleukin-6/biosynthesis , Macrophages/immunology , Monocytes/immunology , RNA-Binding Proteins/immunology , Staphylococcus aureus/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Female , Flow Cytometry , Humans , Intercellular Adhesion Molecule-1/immunology , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/immunology , Lipopolysaccharide Receptors/analysis , Mice , Mice, Inbred BALB C
9.
J R Soc Interface ; 4(14): 533-43, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17251133

ABSTRACT

Upon acute viral infection, a typical cytotoxic T lymphocyte (CTL) response is characterized by a phase of expansion and contraction after which it settles at a relatively stable memory level. Recently, experimental data from mice infected with murine cytomegalovirus (MCMV) showed different and unusual dynamics. After acute infection had resolved, some antigen specific CTL started to expand over time despite the fact that no replicative virus was detectable. This phenomenon has been termed as "CTL memory inflation". In order to examine the dynamics of this system further, we developed a mathematical model analysing the impact of innate and adaptive immune responses. According to this model, a potentially important contributor to CTL inflation is competition between the specific CTL response and an innate natural killer (NK) cell response. Inflation occurs most readily if the NK cell response is more efficient than the CTL at reducing virus load during acute infection, but thereafter maintains a chronic virus load which is sufficient to induce CTL proliferation. The model further suggests that weaker NK cell mediated protection can correlate with more pronounced CTL inflation dynamics over time. We present experimental data from mice infected with MCMV which are consistent with the theoretical predictions. This model provides valuable information and may help to explain the inflation of CMV specific CD8+T cells seen in humans as they age.


Subject(s)
Models, Immunological , T-Lymphocytes, Cytotoxic/immunology , Virus Diseases/immunology , Animals , Herpesviridae Infections/immunology , Humans , Immunologic Memory , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Muromegalovirus
10.
Immun Ageing ; 3: 11, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17156440

ABSTRACT

BACKGROUND: Cellular immunity plays a crucial role in cytomegalovirus (CMV) infection and substantial populations of CMV-specific T cells accumulate throughout life. However, although CMV infection occurs during childhood, relatively little is know about the typical quantity and quality of T cell responses in pediatric populations. METHODS: One thousand and thirty-six people (Male/Female = 594/442, Age: 0-19 yr.; 959 subjects, 20-29 yr.; 77 subjects) were examined for HLA typing. All of 1036 subjects were tested for HLA-A2 antigen. Of 1036 subjects, 887 were also tested for HLA-A23, 24 antigens. In addition, 50 elderly people (Male/Female = 11/39, Age: 60-92 yr.) were also tested for HLA-A2 antigen. We analyzed the CD8+ T cell responses to CMV, comparing these to responses in children and young. The frequencies, phenotype and function CD8+ T cells for two imunodominant epitopes from pp65 were measured. RESULTS: We observed consistently high frequency and phenotypically "mature" (CD27 low, CD28 low, CD45RA+) CMV-specific CD8+ T cell responses in children, including those studied in the first year of life. These CD8+ T cells retained functionality across all age groups, and showed evidence of memory "inflation" only in later adult life. CONCLUSION: CMV consistently elicits a very strong CD8+ T cell response in infants and large pools of CMV specific CD8+ T cells are maintained throughout childhood. The presence of CMV may considerably mould the CD8+ T cell compartment over time, but the relative frequencies of CMV-specific cells do not show the evidence of a population-level increase during childhood and adulthood. This contrast with the marked expansion ("inflation") of such CD8+ T cells in older adults. This study indicates that large scale analysis of peptide specific T cell responses in infants is readily possible. The robust nature of the responses observed suggests vaccine strategies aimed at priming and boosting CD8+ T cells against major pathogens (including HIV, malaria and CMV itself) could be successful in this age-group.

11.
J Immunol ; 177(1): 450-8, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785542

ABSTRACT

CMVs are beta herpesviruses that establish lifelong latent infection of their hosts. Acute infection of C57BL/6 mice with murine CMV elicits a very broad CD8 T cell response, comprising at least 24 epitopes from 18 viral proteins. In contrast, we show here that the CD8 T cell response in chronically infected mice was dominated by only five epitopes. Altogether, four distinct CD8 T cell kinetic patterns were evident. Responses to some epitopes, including M45, which dominates the acute response, contracted sharply after day 7 and developed into stable long-term memory. The response to m139 underwent rapid expansion and contraction, followed by a phase of memory inflation, whereas the response to an M38 epitope did not display any contraction phase. Finally, responses against two epitopes encoded by the immediate early gene IE3 were readily detectable in chronically infected mice but near the limit of detection during acute infection. CD8 T cells specific for the noninflationary M45 epitope displayed a classic central memory phenotype, re-expressing the lymph node homing receptor CD62L and homeostatic cytokine receptors for IL-7 and IL-15, and produced low levels of IL-2. Responses to two inflationary epitopes, m139 and IE3, retained an effector memory surface phenotype (CD62L(low), IL-7Ralpha(-), IL-15Rbeta(-)) and were unable to produce IL-2. We suggest that immunological choices are superimposed on altered viral gene expression profiles to determine immunodominance during chronic murine CMV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Herpesviridae Infections/immunology , Immunologic Memory , Muromegalovirus/immunology , 3T3 Cells , Acute Disease , Amino Acid Sequence , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Proliferation , Chronic Disease , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Growth Inhibitors/administration & dosage , Growth Inhibitors/immunology , Growth Inhibitors/metabolism , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Immediate-Early Proteins/administration & dosage , Immediate-Early Proteins/immunology , Immediate-Early Proteins/metabolism , Immunodominant Epitopes , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/immunology
12.
Eur J Immunol ; 35(11): 3131-41, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16220536

ABSTRACT

In humans, loss of CD27 expression is associated with the stable acquisition of effector functions by CD8+ T cells. We found that murine (CD8+)CD27- T cells were confined to the primed CD62L(dull/-)CD44(bright)CCR7- T cell population. (CD8+)CD27- T cells were absent from lymph nodes but could be found in blood, spleen and in non-lymphoid organs such as lung and liver. Late after primary influenza virus infection, low percentages of antigen-specific CD27- cells emerged in the lung and spleen. After recovery from secondary influenza virus infection, high percentages of influenza-specific CD27- T cells were found in the lung and the loss of CD27 on lung CD8+ T cells coincided with high granzyme B expression. After murine cytomegalovirus infection, loss of CD27 expression on virus-specific CD8+ T cell populations was sustained and especially marked in liver and lung. We suggest that in mice, CD27 is lost from CD8+ T cells only after repetitive antigenic stimulation. Moreover, the high expression of both granzyme B and perforin in the CD27- T cells suggests that the lack of CD27 on murine CD8+ T cells can be used to identify memory T cells with expression of cytotoxic effector molecules.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Aging/blood , Aging/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cytomegalovirus , Cytomegalovirus Infections/immunology , Granzymes , Immunologic Memory , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Perforin , Pore Forming Cytotoxic Proteins , Serine Endopeptidases/metabolism , T-Lymphocyte Subsets/metabolism
13.
Eur J Immunol ; 35(4): 1113-23, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15756645

ABSTRACT

Cytomegalovirus (CMV) is a major human pathogen normally controlled by cellular immune responses. The infection can be modeled in the mouse using murine CMV (MCMV). During the latent phase of infection, two different patterns of CD8(+) T cell responses have been observed: some specificities show increasing frequencies over time ("memory inflation"), while others, which are present acutely, are barely detectable at later time points. This distinction is independent of initial immunodominance. We analyzed the extent to which such responses differ functionally and tracked both their population distribution and their evolution over time. We observed two clear patterns of memory development that diverged early after infection. Acutely, CD8(+) T cells directed against all epitopes showed similar activation, phenotype and distribution. Thereafter, one set of responses ("inflationary") increased in frequency over time, was found in high numbers in non-lymphoid organs and was associated with an activated (CD28(low) CD27(low)CD122(low)) phenotype. In contrast, CD8(+) T cells responses specific for other MCMV epitopes ("non-inflationary") showed a slow reversion to a classical "central" memory phenotype without enrichment in non-lymphoid organs. A simple model to describe the equilibrium state in MCMV is presented, which may point to previously unexplored antiviral populations present after human CMV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Muromegalovirus/immunology , Animals , Antibody Specificity , Antigens/immunology , Kinetics , Mice
14.
Eur J Immunol ; 34(6): 1526-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15162421

ABSTRACT

The frequency and phenotype of human antiviral memory CD8(+) T cells in blood are well studied, yet little is known about their distribution within tissues. Analysis of antiviral CD8(+) T cell populations derived from a unique set of normal liver and blood samples identified a consistent population of virus-specific cells within the liver. In comparison to the circulating T cells, the liver-derived T cells were present at frequencies which were variably enriched compared to that in the blood, and showed significant differences with regard to the expression of CD45RA, CD45RO, CD95, CCR7, CD27 and CD28. The differences in these cell surface markers are consistent with a mature 'effector memory' phenotype of antigen-specific CD8(+) T cells within the liver. An enrichment of an activated subset of NKT cells (V alpha 24/V beta 11) was also observed, a finding which may be relevant to the regulation of the antiviral populations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Liver/immunology , Animals , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/virology , Cytomegalovirus Infections/blood , Epstein-Barr Virus Infections/blood , Flow Cytometry , Humans , Immunologic Memory/immunology , Immunophenotyping , Liver/virology , Mice
15.
J Virol ; 78(5): 2255-64, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14963122

ABSTRACT

CD8(+) T cells are critical for the control of many persistent viral infections, such as human immunodeficiency virus, hepatitis C virus, Epstein-Barr virus, and cytomegalovirus (CMV). In most infections, large CD8(+)-T-cell populations are induced early but then contract and are maintained thereafter at lower levels. In contrast, CD8(+) T cells specific for murine CMV (MCMV) have been shown to gradually accumulate after resolution of primary infection. This unique behavior is restricted to certain epitopes, including an immunodominant epitope derived from the immediate-early 1 (IE1) gene product. To explore the mechanism behind this further, we measured CD8(+)-T-cell-mediated immunity induced by recombinant MCMV-expressing epitopes derived from influenza A virus or lymphocytic choriomeningitis virus placed under the control of an IE promoter. We observed that virus-specific CD8(+)-T-cell populations were induced and that these expanded gradually over time. Importantly, these CD8(+) T cells provided long-term protection against challenge without boosting. These results demonstrate a unique pattern of accumulating T cells, which provide long-lasting immune protection, that is independent of the initial immunodominance of the epitope and indicates the potential of T-cell-inducing vaccines based on persistent vectors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Viral Proteins , Animals , CD8-Positive T-Lymphocytes/cytology , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Herpesviridae Infections/immunology , Immediate-Early Proteins/immunology , Immunodominant Epitopes/immunology , Mice , Mice, Inbred C57BL , Muromegalovirus/genetics , Muromegalovirus/immunology , Muromegalovirus/physiology , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/immunology
16.
J Immunol ; 170(4): 2022-9, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574372

ABSTRACT

CD8+ T lymphocytes play an important role in the control of intracellular pathogens during both acute and persistent infections. This is particularly true in the case of persistent herpesviruses such as human CMV, which are typified by large virus-specific CD8+ T cell populations during viral latency. To understand the origin of these populations and the factors shaping them over time, we investigated the CD8+ T cell response after murine CMV (MCMV) infection. The kinetics of the acute response were characterized by rapid expansion of activated T cells, followed by a contraction phase. Thereafter, we observed a striking pattern, where MCMV-specific memory CD8+ T cells steadily accumulated over time, with 20% of all CD8+ T cells at 1 year specific for one MCMV epitope. Accumulation of MCMV-specific CD8+ T lymphocytes was seen in all organs tested and was associated with continuous activation of specific CD8+ T lymphocytes, primarily within lymph nodes. The pattern of accumulation was observed in only two of five epitopes tested, and was accompanied by a gradual restriction in usage of the variable region of the TCR beta-chain over time. This novel pattern of a virus-specific CD8+ T cell response suggests that continuous or repetitive exposure to Ag can slowly mold memory T cell populations over time. This may be relevant for understanding the evolution of the large human CMV-specific CD8+ T cell populations seen in humans.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Immunologic Memory , Muromegalovirus/immunology , Virus Latency/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Division/immunology , Clone Cells , Epitopes, T-Lymphocyte/immunology , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immediate-Early Proteins/immunology , Immunophenotyping , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred BALB C , Muromegalovirus/physiology , Organ Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , Vaccinia/immunology , Vaccinia/virology , Vaccinia virus/immunology , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...