Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Math Biol ; 88(6): 71, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38668894

ABSTRACT

In epidemics, waning immunity is common after infection or vaccination of individuals. Immunity levels are highly heterogeneous and dynamic. This work presents an immuno-epidemiological model that captures the fundamental dynamic features of immunity acquisition and wane after infection or vaccination and analyzes mathematically its dynamical properties. The model consists of a system of first order partial differential equations, involving nonlinear integral terms and different transfer velocities. Structurally, the equation may be interpreted as a Fokker-Planck equation for a piecewise deterministic process. However, unlike the usual models, our equation involves nonlocal effects, representing the infectivity of the whole environment. This, together with the presence of different transfer velocities, makes the proved existence of a solution novel and nontrivial. In addition, the asymptotic behavior of the model is analyzed based on the obtained qualitative properties of the solution. An optimal control problem with objective function including the total number of deaths and costs of vaccination is explored. Numerical results describe the dynamic relationship between contact rates and optimal solutions. The approach can contribute to the understanding of the dynamics of immune responses at population level and may guide public health policies.


Subject(s)
Communicable Diseases , Mathematical Concepts , Models, Immunological , Vaccination , Humans , Vaccination/statistics & numerical data , Communicable Diseases/immunology , Communicable Diseases/epidemiology , Communicable Diseases/transmission , Computer Simulation , Epidemics/statistics & numerical data , Epidemiological Models
2.
Cent Eur J Oper Res ; 31(2): 499-521, 2023.
Article in English | MEDLINE | ID: mdl-36105892

ABSTRACT

Optimal distribution of vaccines to achieve high population immunity levels is a desirable aim in infectious disease epidemiology. A distributed optimal control epidemiological model that accounts for vaccination was developed and applied to the case of the COVID-19 pandemic. The model proposed here is nonstandard and takes into account the heterogeneity of the infected sub-population with respect to the time since infection, which is essential in the case of COVID-19. Based on the epidemiological characteristics of COVID-19 we analyze several vaccination scenarios and an optimal vaccination policy. In particular we consider random vaccination over the whole population and the prioritization of age groups such as the elderly and compare the effects with the optimal solution. Numerical results of the model show that random vaccination is efficient in reducing the overall number of infected individuals. Prioritization of the elderly leads to lower mortality though. The optimal strategy in terms of total deaths is early prioritization of those groups having the highest contact rates.

3.
Cancer Cell ; 35(4): 649-663.e10, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30991025

ABSTRACT

The BCR-ABL1 fusion protein is the cause of chronic myeloid leukemia (CML) and of a significant fraction of adult-onset B cell acute lymphoblastic leukemia (B-ALL) cases. Using mouse models and patient-derived samples, we identified an essential role for γ-catenin in the initiation and maintenance of BCR-ABL1+ B-ALL but not CML. The selectivity was explained by a partial γ-catenin dependence of MYC expression together with the susceptibility of B-ALL, but not CML, to reduced MYC levels. MYC and γ-catenin enabled B-ALL maintenance by augmenting BIRC5 and enforced BIRC5 expression overcame γ-catenin loss. Since γ-catenin was dispensable for normal hematopoiesis, these lineage- and disease-specific features of canonical Wnt signaling identified a potential therapeutic target for the treatment of BCR-ABL1+ B-ALL.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Wnt Signaling Pathway , gamma Catenin/metabolism , Animals , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Survivin/genetics , Survivin/metabolism , beta Catenin/genetics , beta Catenin/metabolism , gamma Catenin/genetics
4.
J Immunol ; 193(6): 2784-91, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25127860

ABSTRACT

Protection against reinfection is mediated by Ag-specific memory CD8 T cells, which display stem cell-like function. Because canonical Wnt (Wingless/Int1) signals critically regulate renewal versus differentiation of adult stem cells, we evaluated Wnt signal transduction in CD8 T cells during an immune response to acute infection with lymphocytic choriomeningitis virus. Whereas naive CD8 T cells efficiently transduced Wnt signals, at the peak of the primary response to infection only a fraction of effector T cells retained signal transduction and the majority displayed strongly reduced Wnt activity. Reduced Wnt signaling was in part due to the downregulation of Tcf-1, one of the nuclear effectors of the pathway, and coincided with progress toward terminal differentiation. However, the correlation between low and high Wnt levels with short-lived and memory precursor effector cells, respectively, was incomplete. Adoptive transfer studies showed that low and high Wnt signaling did not influence cell survival but that Wnt high effectors yielded memory cells with enhanced proliferative potential and stronger protective capacity. Likewise, following adoptive transfer and rechallenge, memory cells with high Wnt levels displayed increased recall expansion, compared with memory cells with low Wnt signaling, which were preferentially effector-like memory cells, including tissue-resident memory cells. Thus, canonical Wnt signaling identifies CD8 T cells with enhanced proliferative potential in part independent of commonly used cell surface markers to discriminate effector and memory T cell subpopulations. Interventions that maintain Wnt signaling may thus improve the formation of functional CD8 T cell memory during vaccination.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Wnt Proteins/immunology , Wnt Signaling Pathway/immunology , Adoptive Transfer , Animals , Axin Protein/biosynthesis , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Proliferation , Down-Regulation , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Immunologic Memory/immunology , Lectins, C-Type , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/biosynthesis , T-Lymphocyte Subsets/immunology , Vaccination
5.
J Immunol ; 191(10): 5044-51, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24098052

ABSTRACT

Although NK cells use invariant receptors to identify diseased cells, they nevertheless adapt to their environment, including the presence of certain MHC class I (MHC-I) molecules. This NK cell education, which is mediated by inhibitory receptors specific for MHC-I molecules, changes the responsiveness of activating NK cell receptors (licensing) and modifies the repertoire of MHC-I receptors used by NK cells. The fact that certain MHC-I receptors have the unusual capacity to recognize MHC-I molecules expressed by other cells (trans) and by the NK cell itself (cis) has raised the question regarding possible contributions of the two types of interactions to NK cell education. Although the analysis of an MHC-I receptor variant suggested a role for cis interaction for NK cell licensing, adoptive NK cell transfer experiments supported a key role for trans recognition. To reconcile some of these findings, we have analyzed the impact of cell type-specific deletion of an MHC-I molecule and of a novel MHC-I receptor variant on the education of murine NK cells when these mature under steady-state conditions in vivo. We find that MHC-I expression by NK cells (cis) and by T cells (trans), and MHC-I recognition in cis and in trans, are both needed for NK cell licensing. Unexpectedly, modifications of the MHC-I receptor repertoire are chiefly dependent on cis binding, which provides additional support for an essential role for this unconventional type of interaction for NK cell education. These data suggest that two separate functions of MHC-I receptors are needed to adapt NK cells to self-MHC-I.


Subject(s)
H-2 Antigens/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Adoptive Transfer , Animals , Cell Line , H-2 Antigens/genetics , Histocompatibility Antigens Class I/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/immunology
6.
Nat Immunol ; 13(6): 569-78, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22544394

ABSTRACT

Clonal deletion of autoreactive thymocytes is important for self-tolerance, but the intrathymic signals that induce clonal deletion have not been clearly identified. We now report that clonal deletion during negative selection required CD28-mediated costimulation of autoreactive thymocytes at the CD4(+)CD8(lo) intermediate stage of differentiation. Autoreactive thymocytes were prevented from undergoing clonal deletion by either a lack of CD28 costimulation or transgenic overexpression of the antiapoptotic factors Bcl-2 or Mcl-1, with surviving thymocytes differentiating into anergic CD4(-)CD8(-) double-negative thymocytes positive for the T cell antigen receptor αß subtype (TCRαß) that 'preferentially' migrated to the intestine, where they re-expressed CD8α and were sequestered as CD8αα(+) intraepithelial lymphocytes (IELs). Our study identifies costimulation by CD28 as the intrathymic signal required for clonal deletion and identifies CD8αα(+) IELs as the developmental fate of autoreactive thymocytes that survive negative selection.


Subject(s)
Cell Differentiation/immunology , Clonal Deletion/immunology , Receptors, Antigen, T-Cell/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , CD28 Antigens/immunology , CD4 Antigens/immunology , CD8 Antigens/immunology , Flow Cytometry , Immune Tolerance/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , Thymocytes/cytology , Thymus Gland/cytology
7.
Curr Opin Immunol ; 23(2): 228-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21159498

ABSTRACT

The effector response of natural killer (NK) cells is determined by opposing signals received through activating and inhibitory receptors. A process termed NK cell education, which is guided by the recognition of Major Histocompatibility Complex class I (MHC-I) molecules, determines how efficiently activating receptors respond to stimulation. This ensures NK cell tolerance to healthy tissues while allowing robust responses to diseased host cells. It was thought that NK cells are educated during their development in the bone marrow and that education fixes the NK cells' functional properties. However, recent findings suggest that the function of mature peripheral NK cells can adapt to changes in their environment and that the persistent exposure to normal-self is essential to maintain NK cell reactivity. Notwithstanding, NK cell stimulation in the context of inflammation can stably improve the functional properties of NK cells.


Subject(s)
Immunologic Memory , Killer Cells, Natural/immunology , Adaptive Immunity , Cell Differentiation , Histocompatibility Antigens Class I/immunology , Humans , Immune Tolerance , Killer Cells, Natural/cytology
8.
Front Immunol ; 2: 55, 2011.
Article in English | MEDLINE | ID: mdl-22566845

ABSTRACT

Mouse natural killer (NK) cells express Ly49 family receptors that recognize major histocompatibility complex class I (MHC-I) molecules. By interacting with MHC-I molecules expressed on other cells (in trans), inhibitory Ly49 receptors prevent the NK cell-mediated killing of normal cells. In addition, some Ly49 receptors have the unusual property to also interact with MHC-I molecules expressed by the NK cell itself (in cis). cis Binding sequesters a significant fraction of the NK cells' Ly49 receptors, reducing the number of receptors available for trans binding. This lowers the threshold at which NK cell activation exceeds inhibition rendering NK cells more sensitive. It is unclear how Ly49 receptors can bind MHC-I in trans and in cis using the same binding site. We have proposed that this is mediated by two distinct conformations of Ly49 receptors. Here we have tested this model by inferring the distance between the ligand-binding domain of Ly49A and the cell membrane using fluorescence resonance energy transfer (FRET). Consistent with the concept, reducing the distance between the ligand-binding domain of Ly49A and the cell membrane, by shortening the Ly49A stalk, resulted in a substantially increased FRET. The co-expression of cognate MHC-I ligand reduced FRET derived from Ly49A variants with a shortened stalk, indicating that cis association alters FRET. Indeed, FRET improved when cis complexes were disrupted using acid-mediated destruction of MHC-I complexes. These data provide direct evidence that the interaction with MHC-I in cis is associated with a conformational change in the Ly49A receptor on the surface of live cells. The novel FRET based approach may be generally applicable to study conformational changes in cell surface receptors.

9.
J Immunol ; 182(6): 3846-54, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265164

ABSTRACT

Most memory CD8 T cell subsets that have been hitherto defined are generated in response to infectious pathogens. In this study, we have characterized the CD8 T cells that survive priming conditions, devoid of pathogen-derived danger signals. In both a TCR-transgenic model and a model of contact hypersensitivity, we show that the priming of naive CD8 T cells under sterile inflammatory conditions generates memory. The corresponding memory CD8 T cells can be identified by their intermediate expression levels of CD44 and CD122. We also show that CD44/122(int) memory CD8 T cells spontaneously develop in wild type mice and that they display intermediate levels of several other memory traits including functional (IFN-gamma secretion capacity, CCL5 messenger stores), phenotypic, and molecular (T-bet and eomesodermin expression levels) features. We finally show that they correspond to an early differentiation stage and can further differentiate in CD44/122(high) memory T cells. Altogether, our results identify a new memory CD8 T cell subset that is generated under sterile inflammatory conditions and involved in the recall contact hypersensitivity reactions that are responsible for allergic contact dermatitis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Hyaluronan Receptors/physiology , Immunologic Memory/genetics , Inflammation Mediators/physiology , Interleukin-2 Receptor beta Subunit/physiology , Lymphocyte Activation/immunology , Animals , Biomarkers/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Hyaluronan Receptors/biosynthesis , Inflammation Mediators/metabolism , Interleukin-2 Receptor beta Subunit/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
10.
Int Immunol ; 21(2): 123-35, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19088062

ABSTRACT

It has been shown previously that CD8beta in vitro increases the range and the sensitivity of antigen recognition and in vivo plays an important role in the thymic selection of CD8+ T cells. Consistent with this, we report here that CD8+ T cells from CD8beta knockout (KO) P14 TCR transgenic mice proliferate inefficiently in vitro. In contrast to these findings, we also show that CD8beta KO mice mount normal CD8 primary, secondary and memory responses to acute infection with lymphocytic choriomeningitis virus. Tetramer staining and cytotoxic experiments revealed a predominance of CD8-independent CTL in CD8beta KO mice. The TCR repertoire, especially the one of the TCRalpha chain, was different in CD8beta KO mice as compared with B6 mice. Our results indicate that in the absence of CD8beta, CD8-independent TCRs are preferentially selected, which in vivo effectively compensates for the reduced co-receptor function of CD8alphaalpha.


Subject(s)
Antigens, Viral/metabolism , Arenaviridae Infections/immunology , CD8 Antigens/genetics , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocytic choriomeningitis virus/immunology , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Transfection
11.
J Biol Chem ; 283(35): 24254-63, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18579518

ABSTRACT

CD8(+) cytotoxic T lymphocytes (CTL) can recognize and kill target cells expressing only a few cognate major histocompatibility complex (MHC) I-peptide complexes. This high sensitivity requires efficient scanning of a vast number of highly diverse MHC I-peptide complexes by the T cell receptor in the contact site of transient conjugates formed mainly by nonspecific interactions of ICAM-1 and LFA-1. Tracking of single H-2K(d) molecules loaded with fluorescent peptides on target cells and nascent conjugates with CTL showed dynamic transitions between states of free diffusion and immobility. The immobilizations were explained by association of MHC I-peptide complexes with ICAM-1 and strongly increased their local concentration in cell adhesion sites and hence their scanning by T cell receptor. In nascent immunological synapses cognate complexes became immobile, whereas noncognate ones diffused out again. Interfering with this mobility modulation-based concentration and sorting of MHC I-peptide complexes strongly impaired the sensitivity of antigen recognition by CTL, demonstrating that it constitutes a new basic aspect of antigen presentation by MHC I molecules.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , H-2 Antigens/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , H-2 Antigens/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , L Cells , Mice , Peptides/genetics , Receptors, Antigen, T-Cell/genetics
12.
J Immunol ; 177(6): 3903-12, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16951353

ABSTRACT

The isolation of subsets of Ag-specific T cells for in vitro and in vivo studies by FACS is compromised by the fact that the soluble MHC-peptide complexes and Abs used for staining, especially when combined, induce unwanted T cell activation and eventually apoptosis. This is especially a problem for CD8+ CTL, which are susceptible to activation-dependent cell death. In this study, we show that reversible MHC-peptide complexes (tetramers) can be prepared by conjugating MHC-peptide monomers with desthiobiotin (DTB; also called dethiobiotin) and multimerization by reaction with fluorescent streptavidin. While in the cold these reagents are stable and allow good staining, they rapidly dissociate in monomers at elevated temperatures, especially in the presence of free biotin. FACS cloning of Melan-A (MART-1)-specific CTL from a melanoma-infiltrated lymph node with reversible HLA-A2 Melan-A26-35 multimers yielded over two times more clones than when using the conventional biotin-containing multimers. CTL clones obtained by means of reversible multimers killed Melan-A-positive tumor cells more efficiently as compared with clones obtained with the stable multimers. Among the CTL obtained with the reversible multimers, but much less among those obtained with the stable multimers, a high proportion of clones exhibited high functional and physical avidity and died upon incubation with soluble MHC-peptide complexes. Finally, we show that Fab' of an anti-CD8 Ab can be converted in reversible DTB streptavidin conjugates the same way. These DTB reagents efficiently and reversibly stained murine and human CTL without affecting their viability.


Subject(s)
Flow Cytometry , Histocompatibility Antigens Class I , Immunoglobulin Fab Fragments , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Animals , Biotin/analogs & derivatives , Cell Separation , Clone Cells , H-2 Antigens , HLA-A Antigens , HLA-A2 Antigen , Humans , Mice , Peptides , T-Lymphocytes, Cytotoxic/chemistry
13.
J Immunol ; 176(6): 3356-65, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16517703

ABSTRACT

Soluble MHC-peptide (pMHC) complexes induce intracellular calcium mobilization, diverse phosphorylation events, and death of CD8+ CTL, given that they are at least dimeric and co-engage CD8. By testing dimeric, tetrameric, and octameric pMHC complexes containing spacers of different lengths, we show that their ability to activate CTL decreases as the distance between their subunit MHC complexes increases. Remarkably, pMHC complexes containing long rigid polyproline spacers (> or =80 A) inhibit target cell killing by cloned S14 CTL in a dose- and valence-dependent manner. Long octameric pMHC complexes abolished target cell lysis, even very strong lysis, at nanomolar concentrations. By contrast, an altered peptide ligand antagonist was only weakly inhibitory and only at high concentrations. Long D(b)-gp33 complexes strongly and specifically inhibited the D(b)-restricted lymphocytic choriomeningitis virus CTL response in vitro and in vivo. We show that complications related to transfer of peptide from soluble to cell-associated MHC molecules can be circumvented by using covalent pMHC complexes. Long pMHC complexes efficiently inhibited CTL target cell conjugate formation by interfering with TCR-mediated activation of LFA-1. Such reagents provide a new and powerful means to inhibit Ag-specific CTL responses and hence should be useful to blunt autoimmune disorders such as diabetes type I.


Subject(s)
Cytotoxicity, Immunologic/immunology , Histocompatibility Antigens/immunology , Histocompatibility Antigens/metabolism , Peptide Fragments/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Calcium/metabolism , Cell Adhesion , Cell Death , Cells, Cultured , Cross-Linking Reagents/chemistry , Dimerization , Histocompatibility Antigens/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Peptide Fragments/chemistry , Peptides/chemistry , Protein Binding , Solubility , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism
14.
Mol Cell Biol ; 26(3): 735-42, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428431

ABSTRACT

Activation of interleukin-1 (IL-1) receptor (IL-1R), Toll-like receptor 2 (TLR2), and TLR4 triggers NF-kappaB and mitogen-activated protein kinase (MAPK)-dependent signaling, thereby initiating immune responses. Tollip has been implicated as a negative regulator of NF-kappaB signaling triggered by these receptors in in vitro studies. Here, deficient mice were used to determine the physiological contribution of Tollip to immunity. NF-kappaB, as well as MAPK, signaling appeared normal in Tollip-deficient cells stimulated with IL-1beta or the TLR4 ligand lipopolysaccharide (LPS). Similarly, IL-1beta- and TLR-driven activation of dendritic cells and lymphocytes was indistinguishable from wild-type cells. In contrast, the production of the proinflammatory cytokines, IL-6 and tumor necrosis factor alpha was significantly reduced after IL-1beta and LPS treatment at low doses but not at lethal doses of LPS. Tollip therefore controls the magnitude of inflammatory cytokine production in response to IL-1beta and LPS.


Subject(s)
Dendritic Cells/immunology , Interleukin-1/pharmacology , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lipopolysaccharides/pharmacology , Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Dendritic Cells/drug effects , Down-Regulation , Interleukin-1 Receptor-Associated Kinases , Interleukin-6/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocyte Activation/genetics , Lymphocytes/drug effects , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/genetics
15.
J Immunol ; 175(1): 189-95, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972647

ABSTRACT

Mature dendritic cells (DCs) have the capacity to induce efficient primary T cell response and effector cell differentiation. Thus, these cells are a major tool in the design of various immunotherapeutic protocols. We have tested the capacity of different subsets of matured DCs pulsed with a peptide to induce the differentiation of naive CD8 T cells into memory cells in vivo. Flt3 ligand (FL) induces the differentiation of conventional DCs (cDCs) and plasmacytoid DCs (PDCs) from murine bone marrow precursors in vitro. After maturation, both subsets become strong stimulators of Ag-specific T cell responses in vitro. However, the in vivo T cell stimulatory capacity of these DC subsets has not been studied in detail. In the present study, we demonstrate that mature FL-generated DCs induce efficient peptide-specific CD8 T cell response and memory cell differentiation in vivo. This is mainly due to the cDC subset because the PDC subset induced only a negligible primary CD8 response without detectable levels of memory CD8 T cell differentiation. Thus, in vitro FL-generated mature cDCs, but not PDCs, are potent stimulators of peptide-specific CD8 T cell responses and memory generation in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Immunologic Memory/drug effects , Membrane Proteins/pharmacology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Communication/drug effects , Cell Differentiation , Dendritic Cells/classification , Dendritic Cells/cytology , Humans , In Vitro Techniques , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Proteins/pharmacology
16.
J Immunol ; 173(8): 4945-52, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15470036

ABSTRACT

Nucleotide synthesis inhibitors are currently used in neoplastic diseases or as immunosuppressive agents for the prevention of acute rejection in organ transplantation and the treatment of autoimmune disorders. We have previously described that these inhibitors interfere with proliferation and survival of primary T cells in vitro. However, the precise effects of nucleotide restriction on effector and memory functions have not been elucidated. In this study, we investigated the impact of nucleotide synthesis inhibition on CD8 T cell differentiation by using TCR transgenic mice (F5) specific for the influenza virus nucleoprotein 68 peptide presented on the H-2Db molecule. Our results show that methotrexate and 5-fluorouracil prevent the acquisition of effector functions, such as IFN-gamma, granzyme B expression, and cytotoxic function following antigenic stimulation of naive cells. Surprisingly, in the presence of mycophenolate mofetil, activated F5 cells are still able to produce granzyme B and to kill target cells but to a lesser extent compared with control. All three inhibitors interfere with the differentiation of naive cells into memory CD8 T cells. In contrast, the drugs are unable to inhibit the development of improved cytotoxic functions displayed by memory CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Fluorouracil/pharmacology , Immunologic Memory , Methotrexate/pharmacology , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Nucleotides/biosynthesis , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cytotoxicity, Immunologic/drug effects , Graft vs Host Disease/prevention & control , Immunization , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL
17.
J Immunol ; 171(6): 2995-3002, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12960324

ABSTRACT

CpG oligodeoxynucleotide (ODN) promotes maturation of APCs in vivo and induces strong type 1 T cell responses in mice. In this study, we have investigated the ability of CpG1826 to modulate peptide-specific CD8 T cell responses in a context where CD4 T cells are likely to play a minor role. The effects of CpG1826 were evaluated in a system where a population of NP68-specific F5 TCR transgenic CD8 T cells is diluted into a polyclonal host following adoptive transfer into C57BL/10 syngeneic recipients. Using this approach, we found that CpG1826 enhanced the ability of F5 CD8 T cells to undergo multiple divisions in vivo, to express IFN-gamma ex vivo, and to up-regulate memory-associated cell surface markers such as CD122 (IL-2Rbeta) and Ly-6C. Moreover, CpG1826 greatly increased in vivo cytotoxic activity. Using tetramer detection, we found that CpG1826 promoted long-term survival of Ag-specific CD8 T cells after immunization while no NP68-specific cells were detected when the cognate peptide was injected alone. These results indicate that CpG1826 acts as an adjuvant which increases CD8 T cell effector responses and promotes long-term survival of NP68 peptide-specific cells in vivo. They also suggest that this adjuvant can modulate CD8 T cell responses in a system which is likely to be independent of CD4 T cell help.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CpG Islands/immunology , DNA/administration & dosage , Oligodeoxyribonucleotides/administration & dosage , Adoptive Transfer , Animals , Antigens, Ly/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/genetics , Injections, Intraperitoneal , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptides/pharmacology , Perforin , Pore Forming Cytotoxic Proteins , Receptors, Antigen, T-Cell/genetics , Receptors, Interleukin-2/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation/drug effects , Up-Regulation/genetics , Up-Regulation/immunology , fas Receptor/physiology
18.
J Immunol ; 168(5): 2147-53, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11859100

ABSTRACT

Strong memory T cell responses result partly from the selection of Ag-specific clones during immunization. In this study, we show that a monoclonal CD8 T cell population expressing a unique TCR is heterogeneous in terms of clonogenic potential following activation under optimal conditions. More importantly, the frequency of clonogenic cells is strongly increased among Ag-experienced cells, indicating that these cells were either generated or selected during the in vivo primary response. Moreover, strong proliferative responses of primed cells result from this enhanced frequency, as proliferating naive and primed cells display the same cycling parameters, i.e., lag time and intermitotic interval. Hence, these results suggest that the clonogenic potential of individual cells is imprinted before Ag encounter and that clonogenic precursors are selected or generated following in vivo activation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Lymphocyte Activation , Animals , Cells, Cultured , Clone Cells , Genes, RAG-1 , Genes, T-Cell Receptor , Immunization , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/immunology , Stem Cells/immunology , T-Lymphocyte Subsets/immunology , Viral Core Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...