Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
Nat Commun ; 5: 5621, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25470744

ABSTRACT

Psoriasis is a common T-cell-mediated skin disease with 2-3% prevalence worldwide. Psoriasis is considered to be an autoimmune disease, but the precise nature of the autoantigens triggering T-cell activation remains poorly understood. Here we find that two-thirds of patients with moderate-to-severe plaque psoriasis harbour CD4(+) and/or CD8(+) T cells specific for LL37, an antimicrobial peptide (AMP) overexpressed in psoriatic skin and reported to trigger activation of innate immune cells. LL37-specific T cells produce IFN-γ, and CD4(+) T cells also produce Th17 cytokines. LL37-specific T cells can infiltrate lesional skin and may be tracked in patients blood by tetramers staining. Presence of circulating LL37-specific T cells correlates significantly with disease activity, suggesting a contribution to disease pathogenesis. Thus, we uncover a role of LL37 as a T-cell autoantigen in psoriasis and provide evidence for a role of AMPs in both innate and adaptive immune cell activation.


Subject(s)
Autoantigens/immunology , Cathelicidins/immunology , Psoriasis/immunology , T-Lymphocytes/immunology , Antimicrobial Cationic Peptides , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Case-Control Studies , Humans , Immunohistochemistry , Severity of Illness Index
3.
Immunol Lett ; 158(1-2): 134-42, 2014.
Article in English | MEDLINE | ID: mdl-24412596

ABSTRACT

CD28 is a crucial costimulatory receptor necessary full T cell activation. The role of CD28 in multiple sclerosis (MS) has been evaluated as the source of costimulatory signals integrating those delivered by TCR. However, CD28 is also able to act as a unique signaling receptor and to deliver TCR-independent autonomous signals, which regulate the expression and production of pro-inflammatory cytokines and chemokines. By comparing the cytokine/chemokine profiles of CD4(+) T cells from relapsing-remitting multiple sclerosis (RRMS) patients and healthy donors (HD), we found that CD28 engagement without TCR strongly up-regulates IL-8 and IL-6 expression in RRMS compared to HD. More interestingly, in RRMS but not in HD, CD28 stimulation selectively induces the expression of IL-17A by cooperating with IL-6-mediated signals. By using specific inhibitory drugs, we also identify the phosphatidylinositol 3 kinase (PI3K) as the critical regulator of CD28 proinflammatory functions in MS.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Interleukin-17/metabolism , Multiple Sclerosis, Relapsing-Remitting/immunology , Receptors, Antigen, T-Cell/metabolism , Adult , Animals , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Chromones/pharmacology , Female , Humans , Inflammation Mediators/metabolism , Interleukin-17/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , L Cells , Male , Mice , Middle Aged , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Transgenes/genetics , Up-Regulation/drug effects
4.
J Immunol ; 190(10): 5279-86, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23589613

ABSTRACT

CD28 is one of the most relevant costimulatory receptors that deliver both TCR-dependent and TCR-independent signals regulating a wide range of signaling pathways crucial for cytokine and chemokine gene expressions, T cell survival, and proliferation. Most of the CD28-dependent signaling functions are initiated by the recruitment and activation of class IA PI3Ks, which catalyze the conversion of phosphatidylinositol 4,5-biphosphate (PIP2) into phosphatidylinositol 3,4,5-triphosphate, thus generating the docking sites for key signaling proteins. Hence, PIP2 is a crucial substrate in driving the PI3K downstream signaling pathways, and PIP2 turnover may be an essential regulatory step to ensure the activation of PI3K following CD28 engagement. Despite some data evidence that CD28 augments TCR-induced turnover of PIP2, its direct role in regulating PIP2 metabolism has never been assessed. In this study, we show that CD28 regulates PIP2 turnover by recruiting and activating phosphatidylinositol 4-phosphate 5-kinases α (PIP5Kα) in human primary CD4(+) T lymphocytes. This event leads to the neosynthesis of PIP2 and to its consumption by CD28-activated PI3K. We also evidenced that PIP5Kα activation is required for both CD28 unique signals regulating IL-8 gene expression as well as for CD28/TCR-induced Ca(2+) mobilization, NF-AT nuclear translocation, and IL-2 gene transcription. Our findings elucidate a novel mechanism that involves PIP5Kα as a key modulator of CD28 costimulatory signals.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Interleukin-2/genetics , Interleukin-8/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Calcium/metabolism , Cells, Cultured , Enzyme Activation , Gene Expression , Humans , Interleukin-8/genetics , Lymphocyte Activation , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction
5.
PLoS One ; 7(10): e48303, 2012.
Article in English | MEDLINE | ID: mdl-23144749

ABSTRACT

Considerable evidence supports the prediction that CD25 is directly regulated by the forkhead transcription factor FOXP3. However, given that CD25 is normally upregulated in activated T cells, regardless of whether they express FOXP3, this issue has still to be definitively demonstrated. Here we describe that FOXP3, induced by CD28 signals in human CD4(+)CD25(-) T lymphocytes, synergizes with RelA on a regulatory region of Cd25 promoter to mediate the transcriptional activation of Cd25 gene. We found that a striking feature of this regulatory region is the presence of a κB site and of two tandem copies of a non-consensus FOXP3 binding site separated at 5' ends by 19 nucleotides that allow FOXP3 and RelA binding to DNA and their physical interaction. The occupancy of the two FOXP3 binding sites in conjunction with RelA binding site occupancy allows FOXP3 to function as a positive activator of Cd25 gene. Indeed mutations of both FOXP3 binding sites such as mutation of κB site on Cd25 promoter abolished FOXP3 activatory functions. Moreover, FOXP3 mutation ΔE251, that compromises FOXP3 homotypic interactions, failed to trans activate Cd25 promoter, suggesting that both FOXP3 DNA binding and dimerization are required to trans activate Cd25 promoter. These findings identify a novel mechanism by which RelA and FOXP3 cooperate to mediate transcriptional regulation of target genes and characterize a region on Cd25 promoter where FOXP3 dimer could bridge intramolecularly two DNA sites and trans activate Cd25 gene.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Transcription Factor RelA/metabolism , Up-Regulation , Base Sequence , Binding Sites/genetics , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Forkhead Transcription Factors/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Immunoblotting , Interleukin-2 Receptor alpha Subunit/genetics , Jurkat Cells , Lymphocyte Activation , Molecular Sequence Data , Mutation , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/genetics , Transcriptional Activation
6.
PLoS Pathog ; 8(6): e1002759, 2012.
Article in English | MEDLINE | ID: mdl-22737070

ABSTRACT

Caspase-dependent cleavage of antigens associated with apoptotic cells plays a prominent role in the generation of CD8⁺ T cell responses in various infectious diseases. We found that the emergence of a large population of autoreactive CD8⁺ T effector cells specific for apoptotic T cell-associated self-epitopes exceeds the antiviral responses in patients with acute hepatitis C virus infection. Importantly, they endow mixed polyfunctional type-1, type-2 and type-17 responses and correlate with the chronic progression of infection. This evolution is related to the selection of autoreactive CD8⁺ T cells with higher T cell receptor avidity, whereas those with lower avidity undergo prompt contraction in patients who clear infection. These findings demonstrate a previously undescribed strict link between the emergence of high frequencies of mixed autoreactive CD8⁺ T cells producing a broad array of cytokines (IFN-γ, IL-17, IL-4, IL-2…) and the progression toward chronic disease in a human model of acute infection.


Subject(s)
Apoptosis/immunology , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Hepatitis C/immunology , T-Lymphocyte Subsets/immunology , Adult , Disease Progression , Epitopes, T-Lymphocyte/immunology , Female , Hepacivirus/immunology , Humans , Male , Middle Aged , Young Adult
7.
Eur J Immunol ; 41(2): 503-13, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268019

ABSTRACT

The molecular mechanisms whereby CD28 alone or associated with TCR can regulate FOXP3 expression are not understood, although the importance of CD28 as a pivotal regulator of CD4(+) CD25(+) FOXP3(+) T cells is well recognized. We previously demonstrated that unique CD28-induced, NF-κB-dependent signals were sufficient to activate FOXP3 transcription in human CD4(+) CD25(-) T cells; however, the exact mechanisms are currently unknown. In this study, we have identified novel κB-binding sites on FOXP3 gene and demonstrated that CD28 signals mediated FOXP3 trans activation by nuclear translocation of RelA/NF-κB and not of c-Rel. The occupancy of FOXP3 κB-binding sites by RelA dimers that correlated with histone acetylation and recruitment of Pol II were required both to initiate FOXP3 transcription and to control the promoter occupancy by NFAT. Interestingly, knockdown of RelA in CD4(+) CD25(-) T cells stimulated through TCR and CD28 significantly affected FOXP3 expression, confirming that also the transcriptional activation of FOXP3 gene by TCR in the presence of CD28-costimulatory signals is RelA-dependent. In conclusion, these data suggest a new mechanism by which FOXP3 is activated and supports the critical role of CD28 in the regulation of peripheral tolerance.


Subject(s)
CD28 Antigens/immunology , Forkhead Transcription Factors/metabolism , Lymphocyte Activation/physiology , NF-kappa B/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcription Factor RelA/metabolism , Acetylation , Animals , Antibodies/immunology , Antibodies/pharmacology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CD3 Complex/immunology , Cyclosporine/pharmacology , Forkhead Transcription Factors/genetics , HEK293 Cells , Histones/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , L Cells/immunology , L Cells/metabolism , Lymphocyte Activation/drug effects , Mice , NF-kappa B/antagonists & inhibitors , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/genetics , RNA, Small Interfering/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Transfection
8.
Virology ; 386(2): 398-406, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19232664

ABSTRACT

The ability of HCV to mutate in response to cytotoxic T lymphocyte (CTL) pressure is increasingly recognized, but the influence of such a mechanism in viral persistence and final disease outcome has not been ascertained. In this study, we performed a detailed longitudinal analysis of cell mediated immunity and HCV evolution in two self limiting and two chronically evolving HCV acutely infected patients, one of whom transiently controlled viremia. Amino acid mutations in immunodominant regions of viruses were observed in all patients, although they conferred viral escape from CTL responses only in chronically infected individuals. Resurgence of viremia coincided with the replacement of the original virus quasispecies with mutant viruses that had escaped recognition by primary CD8(+) T cell responses and infection persisted in the presence of variant viruses which were less efficiently recognized by preexisting and de novo induced T cell responses.


Subject(s)
Hepacivirus/genetics , Hepatitis C/immunology , Immunity, Cellular , T-Lymphocytes, Cytotoxic/immunology , Adult , Amino Acid Sequence , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Evolution, Molecular , Female , Genome, Viral , Hepacivirus/immunology , Hepatitis C/virology , Humans , Immunodominant Epitopes/immunology , Male , Middle Aged , Molecular Sequence Data , Mutation , Phylogeny , Sequence Alignment , Sequence Analysis, RNA , T-Lymphocytes, Cytotoxic/virology , Viremia/immunology , Viremia/virology
9.
J Immunol ; 181(2): 1025-33, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606654

ABSTRACT

Among the signals necessary to generate CD4(+)CD25(+)FOXP3(+) T cells from CD4(+)CD25(-)FOXP3(-) T cells, a pivotal role is played by CD28. However, in humans, it is not known whether CD28 signaling independently of TCR promotes forkhead box protein 3 (FOXP3) expression and regulates CD4(+)CD25(+)FOXP3(+) T cell functions. To address this issue, starting from our previous experience, we analyzed the unique signals delivered by CD28 following stimulation by its natural ligand B7. Our results show that, in primary CD4(+)CD25(-) T cells, CD28 signals independent of TCR-mediated stimulatory pathways are sufficient to induce the transcription of FOXP3 in a small number of CD4(+)CD25(-) T cells committed to express FOXP3. These signals are dependent on CD28-derived PI3K/Akt pathways and resistant to cyclosporin A. In addition, we demonstrated that translated FOXP3 was recruited to CD25, Il-2, and Ctla4 target promoters. CD28-mediated FOXP3 expression was transient and correlated with CD25 expression. The presence of FOXP3 in CD28-activated CD4(+)CD25(-) T cells correlated with a transient unresponsiveness to antigenic stimuli. The addition of exogenous IL-2 did not influence either FOXP3 or CD25 expression but rescued CD28-activated T cells from apoptosis. Our results, demonstrating that FOXP3 expression driven solely by the CD28/B7 interaction inhibited T cell activation, support the role of CD28 in the regulation of peripheral tolerance and suggest a new mechanism through which it could occur.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Immune Tolerance , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocyte Subsets/immunology , Animals , B7-1 Antigen/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Humans , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/immunology , Mice , T-Lymphocyte Subsets/metabolism
10.
J Hepatol ; 48(2): 216-28, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18180071

ABSTRACT

BACKGROUND/AIMS: Several studies suggest that the evolutionary rate of HVR1 sequence in acute HCV hepatitis derives from the action of a continuous immune-driven positive selection. However, these studies have not been performed examining the relationship between HVR1 evolution and the development of specific immunity to autologous HVR1 sequences. METHODS: We performed a longitudinal analysis of HVR1 sequences and specific antibodies and CD4+ T cells in ten HCV acutely infected patients with different clinical outcomes (recovery versus persistence). RESULTS: We showed that although both recovered and chronically evolving individuals developed IFN-gamma+ T cells specific for Core and NS sequences, HVR1-specific CD4+ T cells were detected only in patients clearing the virus. On the contrary, all patients displayed anti-HVR1 antibodies that recognized sequences exclusively carried by autologous viruses. Measurements of genetic diversity and the number of non-synonymous per synonymous substitutions within HVR1 sequences before and after antibody appearance showed an increase of these parameters only in concomitance with the appearance of anti-HVR1 antibodies. CONCLUSIONS: The evidence that anti-HVR1 antibodies favor HVR1 variant selection suggests that viral complexity in chronically infected patients could represent a virus adaptive strategy to escape the continuous selective process mediated by anti-HVR1 antibodies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepatitis C Antibodies/physiology , Hepatitis C/immunology , Viral Proteins/immunology , Acute Disease , Adult , Amino Acid Sequence , Evolution, Molecular , Female , Genetic Variation , Humans , Longitudinal Studies , Male , Middle Aged , Molecular Sequence Data , Viral Proteins/chemistry , Viral Proteins/genetics
11.
Eur J Immunol ; 35(9): 2627-37, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16114108

ABSTRACT

Cellular immune responses are induced during hepatitis C virus (HCV) infection and acute-phase CD8+ T cells are supposed to play an important role in controlling viral replication. In chimpanzees, failure of CD8+ T cells to control HCV replication has been associated with acquisition of mutations in MHC class I-restricted epitopes. In humans, although selection of escape mutations in an immunodominant CTL epitope has been recently described, the overall impact of immune escape during acute HCV infection is unclear. Here, by performing an in depth analysis of the relationship between early cellular immune responses and viral evolution in a chronically evolving HCV acutely infected individual, we demonstrate: (i) the presence of a potent and focused CD8(+ T cell response against a novel epitope in the NS3 protein, (ii) the elimination of the quasi-species harboring the original amino acid sequence within this epitope, and (iii) the selection for a virus population bearing amino acid changes at a single residue within the cytotoxic T cell epitope that strongly diminished T cell recognition. These results support the view that acute-phase CD8+ T cell responses exert a biologically relevant pressure on HCV replication and that viruses escaping this host response could have a significant survival advantage.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/genetics , Hepacivirus/physiology , Hepatitis C/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Amino Acid Sequence , CD8-Positive T-Lymphocytes/virology , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/immunology , Evolution, Molecular , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis C/virology , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Interferon-gamma/immunology , Male , Molecular Sequence Data , Peptide Fragments/immunology , Sequence Alignment , Sequence Analysis, DNA , T-Lymphocytes, Cytotoxic/virology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Virus Replication/genetics , Virus Replication/immunology
12.
J Leukoc Biol ; 78(2): 372-82, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15923217

ABSTRACT

We have described previously that hypervariable region 1 (HVR1) variants of hepatitis C virus (HCV) frequently act as T cell receptor (TCR) antagonists for HVR1-specific helper T cells. These naturally occurring HVR1-antagonistic sequences interfered with the effects of HVR1-agonistic sequences such as TCR down-regulation and early activatory signals. By taking advantage of these findings, in this paper, we have analyzed the fate of these HVR1-specific antagonized CD4+ T cells. We present the evidence that TCR antagonism renders agonist-activated T cells susceptible to bystander CD95-mediated killing by suppressing the expression of cellular Fas-associated death domain-like interleukin-1beta-converting enzyme-like inhibitor proteins. To verify whether the TCR repertoire of a HVR1-specific T cell population could be modified consequently, we used a HVR1-agonistic sequence to induce in vitro CD4+ T cells and another HVR1 sequence with antagonistic property to mediate suppressive phenomena. HVR1-specific T cells were cultured with the agonist alone or with the agonist plus the antagonist. HVR1 specificity and T cell repertoires were followed over time by analyzing TCR beta-variable gene segment by "spectratyping". The results showed that the specificity for the agonist was rapidly spoiled after culture in the presence of the antagonist, and the TCR repertoire was strongly modified as a result of CD95-mediated apoptosis of agonist-specific clonal expansions. These data support the hypothesis that in HCV infection, the generation of TCR antagonists may reshape the T cell repertoire, representing an efficacious immune evasion strategy of a highly mutant pathogen.


Subject(s)
Antigens, Viral/immunology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Hepacivirus/immunology , Receptors, Antigen, T-Cell, alpha-beta/antagonists & inhibitors , T-Lymphocyte Subsets/immunology , Viral Proteins/immunology , fas Receptor/immunology , Cell Proliferation , Cells, Cultured , Down-Regulation/immunology , Epitopes, T-Lymphocyte , Hepacivirus/pathogenicity , Humans , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Serpins/biosynthesis , Serpins/immunology , Viral Proteins/biosynthesis
13.
Proc Natl Acad Sci U S A ; 101(16): 6098-103, 2004 Apr 20.
Article in English | MEDLINE | ID: mdl-15079071

ABSTRACT

CD28 is one of the most important costimulatory receptors necessary for full T lymphocyte activation. The CD28 receptor can enhance T cell antigen receptor (TCR) signals, as well as deliver independent signals. Indeed, CD28 engagement by B7 can generate TCR-independent signals leading to IkappaB kinase and NF-kappaB activation. Here we demonstrate that the TCR-independent CD28 signal leads to the selective transcription of survival (Bcl-xL) and inflammatory (IL-8 and B cell activation factor, but not proliferative (IL-2), genes, in a NF-kappaB-dependent manner. CD28-stimulated T cells actively secrete IL-8, and Bcl-xL up-regulation protects T cells from radiation-induced apoptosis. The transcription of CD28-induced genes is mediated by the specific recruitment of RelA and p52 NF-kappaB subunits to target promoters. In contrast, p50 and c-Rel, which preferentially bind NF-kappaB sites on the IL-2 gene promoter after anti-CD3 stimulation, are not involved. Thus, we identify CD28 as a key regulator of genes important for both survival and inflammation.


Subject(s)
CD28 Antigens/physiology , Interleukin-8/genetics , NF-kappa B/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/physiology , Base Sequence , Biological Transport , Cell Nucleus/metabolism , DNA Primers , Gene Expression Regulation/physiology , Humans , NF-kappa B/genetics , Transcription Factor RelA , Transcription, Genetic , bcl-X Protein
14.
Microbes Infect ; 6(2): 164-71, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14998514

ABSTRACT

Infection of humans by Toxoplasma gondii leads to an acute systemic phase, in which tachyzoites disseminate throughout the body, followed by a chronic phase characterized by the presence of tissue cysts, containing bradyzoites, in brain, heart and skeletal muscles. This work focused on studying the antigenic regions of bradyzoite-specific proteins involved in human B- and T-cell responses. To this aim, we constructed a phage-display library of DNA fragments derived from the bradyzoite-specific genes BAG1, MAG1, SAG2D, SAG4, BSR4, LDH2, ENO1 and p-ATPase. Challenge of the bradyzoite library with sera of infected individuals led to the identification of antigenic regions within BAG1 and MAG1 gene products. Analysis of the humoral and lymphoproliferative responses to recombinant antigens demonstrated that the BAG1 fragment induced T-cell proliferation in 34% of T. gondii-exposed individuals, while 50% of them had specific IgG. In the same subjects, the MAG1 fragment was recognized by T cells from 17% of the exposed donors and by antibodies from 73% of them. A detailed analysis of the antibody response against BAG1 and MAG1 antigen fragments demonstrated that the immune response against bradyzoites occurs early after infection in humans. Finally, we provide evidence that the T-cell response against BAG1 is associated with the production of interferon-gamma, suggesting that bradyzoite antigens should be considered in the design of potential vaccines in humans.


Subject(s)
Antibody Formation/immunology , Antigens, Protozoan/immunology , Heat-Shock Proteins/immunology , Immunity, Cellular/immunology , Protozoan Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Adult , Amino Acid Sequence , Animals , Cell Culture Techniques , Female , Humans , Molecular Sequence Data , Toxoplasma/genetics , Toxoplasma/physiology
15.
Hepatology ; 38(3): 653-63, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12939592

ABSTRACT

An ideal strategy that leads to a vaccine aimed at controlling viral escape may be that of preventing the replication of escape mutants by eliciting a T- and B-cell repertoire directed against many viral variants. The hypervariable region 1 (HVR1) of the putative envelope 2 protein that presents B and T epitopes shown to induce protective immunity against hepatitis C virus (HCV), might be suitable for this purpose if its immunogenicity can be improved by generating mimics that induce broad, highly cross-reactive, anti-HVR1 responses. Recently we described a successful approach to select HVR1 mimics (mimotopes) incorporating the variability found in a great number of viral variants. In this report we explore whether these mimotopes, designed to mimic B-cell epitopes, also mimic helper T-cell epitopes. The first interesting observation is that mimotopes selected for their reactivity to HVR1-specific antibodies of infected patients also do express HVR1 T-cell epitopes, suggesting that similar constraints govern HVR1-specific humoral and cellular immune responses. Moreover, some HVR1 mimotopes stimulate a multispecific CD4(+) T-cell repertoire that effectively cross-reacts with HVR1 native sequences. This may significantly limit effects as a T-cell receptor (TCR) antagonist frequently exerted by natural HVR1-variants on HVR1-specific T-cell responses. In conclusion, these data lend strong support to using HVR1 mimotopes in vaccines designed to prevent replication of escape mutants.


Subject(s)
Complementarity Determining Regions , Hepacivirus/genetics , Hepatitis C, Chronic/virology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Molecular Mimicry/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Aged , B-Lymphocytes/immunology , Cell Line , Cross Reactions , Epitopes/analysis , Female , Hepatitis C, Chronic/immunology , Humans , Male , Middle Aged , Receptors, Antigen, T-Cell/antagonists & inhibitors , T-Lymphocytes/immunology
16.
Eur J Immunol ; 33(9): 2609-18, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12938237

ABSTRACT

CD4-Lck recruitment to TCR/CD3, as well as Lck activation is essential for T cell activation. Indeed, the blockage of CD4-Lck recruitment to TCR during antigen recognition exerts a drastic inhibitory effect on T cell activation by interfering with both early and late phases of T cell signaling. In the present work, we report a novel inhibitory mechanism by which CD4 can shut down proximal T cell-activating signals. Indeed, we show that upon ligation of CD4 by antibodies the inhibitory kinase, p50(csk), is strongly induced and prolonged during the time. In contrast, p50(csk) was not activated when TCR and CD4 were properly engaged by their ligands. We also demonstrate that anti-CD4 treatment stimulated Csk kinase associated to the membrane adapter, PAG/Cbp, without affecting the total amount of Csk bound to PAG/Cbp. As a consequence, early tyrosine phosphorylation events as well as downstream signaling pathways leading to IL-2 gene expression induced by TCR were inhibited in anti-CD4 pretreated cells. We suggest a new model to explain the activation of negative signals by CD4 molecule.


Subject(s)
CD4 Antigens/metabolism , Enzyme Activation/physiology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , CD4 Antigens/immunology , CSK Tyrosine-Protein Kinase , Enterotoxins/metabolism , Humans , Jurkat Cells , Protein-Tyrosine Kinases/immunology , Signal Transduction/physiology , src-Family Kinases
17.
Eur J Immunol ; 33(8): 2186-96, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12884293

ABSTRACT

Several findings support the importance of GM1-enriched lipid microdomains of plasma membrane and of Vav, an essential regulator of actin cytoskeletal rearrangement, in the regulation of T cell activation. Moreover, a functional link among lipid microdomains, Vav and the HIV product Nef has been described. These observations suggest that Nef can modify plasma membrane GM1, affecting the behavior of HIV-infected cells towards antigen recognition and Vav towards counteracting such an effect. We observed that Nef expression, either following viral infection or ectopic expression, significantly decreased the level of plasma membrane GM1 in unstimulated T cells. This down-regulation was associated with the inhibition of NF-AT activation, but not with NF-kappaB activation induced by TCR engagement. Dissecting the signaling pathway that regulates NF-AT activation, we found that Nef inhibited exclusively the Ca(2+)/calcineurin cascade, whereas the JNK cascade and AP-1 transcriptional activity were not affected. Our evidence that Vav overexpression counteracted both the Nef-induced decrease of GM1 expression and the inhibition of NF-AT activity, suggests a novel mechanism by which Nef may interfere with TCR-mediated activation through the modulation of intracellular trafficking and clustering of GM1-enriched microdomains at the cell surface.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/metabolism , G(M1) Ganglioside/metabolism , Gene Products, nef/metabolism , HIV-1/metabolism , Nuclear Proteins , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Calcium Signaling , Cell Membrane/metabolism , Gene Products, nef/genetics , HIV-1/genetics , Humans , Jurkat Cells , NF-kappa B/metabolism , NFATC Transcription Factors , Proto-Oncogene Proteins c-vav , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transfection , nef Gene Products, Human Immunodeficiency Virus
18.
J Immunol ; 170(6): 2895-903, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12626540

ABSTRACT

We have recently observed that CD28 engagement initiates a signaling pathway leading to the activation of I kappa B kinase (IKK) complex and, consequently, to NF-kappa B activation, and we identified Vav-1 as an important mediator of this function. Here we report for the first time that Vav-1 constitutively associates with IKK alpha in both Jurkat and primary CD4(+) T cells. Vav-1/IKK alpha association is mediated by their helix-loop-helix domains, does not involve IKK beta, and is functionally relevant in that Vav-1-associated IKK alpha kinase activity is increased following CD28 engagement by B7. Moreover, we demonstrate that CD28-induced NF-kappa B activation is augmented by both IKK alpha and Vav-1, but not IKK beta. Confocal microscopy showed that endogenous Vav-1 and IKK alpha, but not IKK beta, were recruited to the membrane and colocalized in response to CD28 stimulation. Taken together, these data evidence that Vav-1 plays a key role in the control of NF-kappa B pathway by targeting IKK alpha in the T cell membrane and favoring its activation in response to CD28 stimulation.


Subject(s)
CD28 Antigens/physiology , Cell Cycle Proteins , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/physiology , Protein Subunits/physiology , Proto-Oncogene Proteins/physiology , Animals , Antibodies, Monoclonal/pharmacology , B7-1 Antigen/pharmacology , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Helix-Loop-Helix Motifs/immunology , Humans , I-kappa B Kinase , Jurkat Cells , L Cells , Lymphocyte Activation , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein Subunits/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-vav , Receptor-CD3 Complex, Antigen, T-Cell/deficiency , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptor-CD3 Complex, Antigen, T-Cell/physiology
19.
J Immunol ; 168(12): 6106-12, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12055221

ABSTRACT

In the present study, we aimed to demonstrate that CD4 may represent a critical turning point that governs the apoptotic and survival programs in T cells, without modifying the physical association with the TCR-CD3 complex. To address this issue, we have explored the possibility that the activation of CD4 may transduce apoptotic signals unless signaling effectors neutralize them. Our data show that in Jurkat T cells CD4 engagement by Leu3a mAb results in a rapid and strong increase of Lck kinase activity, subsequent alterations of mitochondrial membrane potential, and apoptosis. Critical parameters are coassociation of CD4/Lck with TCR/CD3 and up-regulation of the proapoptotic protein Bax. Indeed, Leu3a-mediated Lck activation failed to induce apoptotic features in Jurkat cells either defective for TCR/CD3 or overexpressing the antiapoptotic protein Bcl-2. Furthermore, we demonstrate that Leu3a treatment of Jurkat cells overexpressing Vav results in the inhibition of mitochondrial damage and apoptosis; this rescue effect is accompanied with a significant decrease of Bax expression observed in apoptotic cells. Our evidence that the activation of Lck activates in T cells apoptotic pathways which are counteracted by Vav, a signaling molecule that cooperates with CD28 to boost TCR signals, suggests a novel role for costimulation in protecting T cells from CD4-mediated cell death.


Subject(s)
CD4 Antigens/physiology , Cell Cycle Proteins , Guanine Nucleotide Exchange Factors , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Mitochondria/pathology , Proto-Oncogene Proteins , Proto-Oncogene Proteins/biosynthesis , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Antibodies, Monoclonal/metabolism , Apoptosis/immunology , CD4 Antigens/immunology , CD4 Antigens/metabolism , Down-Regulation/immunology , Enzyme Activation/immunology , Guanine Nucleotide Exchange Factors/biosynthesis , Guanine Nucleotide Exchange Factors/genetics , Humans , Intracellular Membranes/immunology , Intracellular Membranes/pathology , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Membrane Potentials/immunology , Mitochondria/immunology , Permeability , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins c-vav , Signal Transduction/immunology , Transfection , bcl-2-Associated X Protein
20.
J Immunol ; 168(3): 1060-8, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11801639

ABSTRACT

T cell suppression exerted by regulatory T cells represents a well-established phenomenon, but the mechanisms involved are still a matter of debate. Recent data suggest that anergic T cells can suppress responder T cell activation by inhibiting Ag presentation by dendritic cells (DC). In this study, we focused our attention on the mechanisms that regulate the susceptibility of DC to suppressive signals and analyzed the fate of DC and responder T cells. To address this issue, we have cocultured human alloreactive or Ag-specific CD4+ T cell clones, rendered anergic by incubation with immobilized anti-CD3 Ab, with autologous DC and responder T cells. We show that anergic T cells affect either Ag-presenting functions or survival of DC, depending whether immature or mature DC are used as APC. Indeed, MHC and costimulatory molecule expression on immature DC activated by responder T cells is inhibited, while apoptotic programs are induced in mature DC and in turn in responder T cells. Ligation of CD95 by CD95L expressed on anergic T cells in the absence of CD40-CD40L (CD154) interaction are critical parameters in eliciting apoptosis in both DC and responder T cells. In conclusion, these findings indicate that the defective activation of CD40 on DC by CD95L+ CD154-defective anergic T cells could be the primary event in determining T cell suppression and support the role of CD40 signaling in regulating both conditioning and survival of DC.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Clonal Anergy/immunology , Dendritic Cells/immunology , T-Lymphocytes, Regulatory/immunology , Antigens/pharmacology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/metabolism , CD40 Ligand/biosynthesis , Cell Differentiation/immunology , Cells, Cultured , Clonal Deletion , Clone Cells , Coculture Techniques , Dendritic Cells/cytology , Down-Regulation/immunology , Humans , Lymphocyte Activation/immunology , fas Receptor/biosynthesis , fas Receptor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...