Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Lung ; 200(2): 179-185, 2022 04.
Article in English | MEDLINE | ID: mdl-35381867

ABSTRACT

AIMS: The very early management of pulmonary embolism (PE), a part from antithrombotic treatment, has been little studied. Our aim was to compare the effects of diuretic therapy (DT) versus volume expansion (VE) in patients hospitalized for PE with RV dysfunction. METHODS AND RESULTS: We conducted a randomized open-label multicentric study including patients with intermediate high-risk PE. Patients were randomized between diuretics or saline infusion. The primary endpoint was time to troponin (Tp) normalization. Secondary endpoints were time to normalization of B-type natriuretic peptide (BNP), changes in echocardiographic RV function parameters and treatment tolerance. Sixty patients presenting intermediate high-risk PE were randomized. Thirty received DT and 30 VE. We noted no changes in Tp kinetics between the two groups. In contrast, faster normalization of BNP was obtained in the DT group: 56 [28-120] vs 108 [48-144] h: p = 0.05, with a shorter time to 50%-decrease from peak value 36 [24-48] vs 54 [41-67] h, p = 0.003 and a higher rate of patients with a lower BNP concentration within the first 12 h (42% vs 12% p < 0.001). RV echocardiographic parameters were unchanged between the groups. One dose 40 mg furosemide was well-tolerated and not associated with any serious adverse events. CONCLUSION: In the acute management of intermediate high-risk PE, initial therapy including diuretic treatment is well-tolerated and safe. Although changes in Tp kinetics and echocardiographic RV dysfunction parameters did not differ, normalization of BNP is achieved more quickly in the DT group. This finding, which need to be confirmed in trials with clinical end points, may reflects a rapid improvement in RV function using one dose 40 mg furosemide. TRIAL REGISTRY: Clinical Trial Registration NCT02531581.


Subject(s)
Diuretics , Pulmonary Embolism , Ventricular Dysfunction, Right , Acute Disease , Biomarkers , Diuretics/therapeutic use , Furosemide/therapeutic use , Humans , Natriuretic Peptide, Brain , Pulmonary Embolism/complications , Pulmonary Embolism/diagnostic imaging , Pulmonary Embolism/drug therapy , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Dysfunction, Right/drug therapy
2.
BMJ Open ; 11(9): e050910, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588255

ABSTRACT

OBJECTIVES: Severity of a first pulmonary embolism (PE) is sometimes proposed as a criterion for prolonging anticoagulant treatment. However, little evidence supports this idea. We attempted to determine the connection between severity of first PE and the risk of recurrence. PARTICIPANTS: Patients admitted with PE between 2012 and 2018 and for whom anticoagulant treatment had been discontinued were followed. PEs were classified according to the severity into the following two groups: those with associated cardiac involvement (increased cardiac biomarker(s) and/or echocardiographic right ventricular dysfunction) and those with no cardiac involvement which were classified as non-severe. Recurrence-free survivals were estimated using the Kaplan-Meier method and compared using the log-rank test. RESULTS: 417 patients with PEs (186 with cardiac involvement) were followed for at least 1 year after discontinuation of treatment with a mean follow-up of: 3.5±1.9 years. 72 patients (17.3%) experienced venous thromboembolism recurrence: 24 (5.8%), 44 (12 %) and 72 (28.3 %) respectively, at 1, 2 and 5 years. In 63 patients (88%), recurrence was a PE. Mean time to onset of recurrence was 24.9±19.9 months. At 5 years, the recurrence rate is higher when the first PE was associated with cardiac involvement p=0.043. In contrast, in patients with provoked PE, the recurrence rate is higher when the first PE event was associated with cardiac involvement: p=0.032. Multivariate analysis demonstrates that PE severity is an independent factor of recurrence (HR 1.634 (1.015-2.632), p=0.043). CONCLUSION: We report for the first time a possible link between a higher recurrence rate and the severity of the first PE. This result which must be confirmed in a dedicated prospective trial could become an important criterion for the duration of anticoagulant therapy after a PE. TRIAL REGISTRATION NUMBER: NCT04980924.


Subject(s)
Pulmonary Embolism , Venous Thromboembolism , Anticoagulants/therapeutic use , Humans , Prospective Studies , Pulmonary Embolism/drug therapy , Pulmonary Embolism/epidemiology , Recurrence , Retrospective Studies , Risk Factors , Venous Thromboembolism/drug therapy , Venous Thromboembolism/epidemiology
3.
Blood ; 113(2): 347-57, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-18849489

ABSTRACT

By presenting antigenic peptides on the cell surface, human leukocyte antigen (HLA) class I molecules are critical for immune defense. Their surface density determines, to a large extent, the level of CD8(+) T cell-dependent immune reactions; their loss is a major mechanism of immune escape. Therefore, powerful processes should regulate their surface expression. Here we document the mechanisms used by CD99 to mediate HLA class I modulation. Up-regulation of HLA class I by IFN-gamma requires CD99. In the trans Golgi network (TGN), and up to the cell surface, CD99 and HLA class I are physically associated via their transmembrane domain. CD99 also binds p230/golgin-245, a coiled-coil protein that recycles between the cytosol and buds/vesicles of the TGN and which plays a fundamental role in trafficking transport vesicles. p230/golgin-245 is anchored within TGN membranes via its Golgin-97, RanBP1, IMh1p, P230 (GRIP) domain and the overexpression of which leads to surface and intracellular down-modulation of HLA class I molecules.


Subject(s)
Antigens, CD/immunology , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/immunology , Golgi Apparatus/immunology , Histocompatibility Antigens Class I/immunology , Membrane Proteins/immunology , Up-Regulation/immunology , 12E7 Antigen , Antigens, CD/metabolism , Antiviral Agents/immunology , Antiviral Agents/pharmacology , Autoantigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Adhesion Molecules/metabolism , Cytosol/immunology , Cytosol/metabolism , Golgi Apparatus/metabolism , HLA Antigens/biosynthesis , HLA Antigens/immunology , Histocompatibility Antigens Class I/biosynthesis , Humans , Immunity, Cellular/physiology , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Jurkat Cells , Membrane Proteins/metabolism , Protein Structure, Tertiary/physiology , Protein Transport/drug effects , Protein Transport/immunology , Transport Vesicles/immunology , Transport Vesicles/metabolism , Up-Regulation/drug effects
4.
J Immunol ; 180(9): 6054-63, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424726

ABSTRACT

Natural CD25(+)CD4(+) regulatory T cells (Treg) are essential for self-tolerance and for the control of T cell-mediated immune pathologies. However, the identification of Tregs in an ongoing immune response or in inflamed tissues remains elusive. Our experiments indicate that TIRC7, T cell immune response cDNA 7, a novel membrane molecule involved in the regulation of T lymphocyte activation, identifies two Treg subsets (CD25(low)TIRC7(+) and CD25(high)TIRC7(-)) that are characterized by the expression of Foxp3 and a suppressive activity in vitro and in vivo. We also showed that the CD25(low)TIRC7(+) subset represents IL-10-secreting Tregs in steady state, which is accumulated intratumorally in a tumor-bearing mice model. Blockade of the effect of IL-10 reversed the suppression imposed by the CD25(low)TIRC7(+) subset. Interestingly, these IL-10-secreting cells derived from the CD25(high)TIRC7(-) subset, both in vitro and in vivo, in response to tumoral Ags. Our present results strongly support the notion that, in the pool of natural Tregs, some cells can recognize foreign Ags and that this recognition is an essential step in their expansion and suppressive activity in vivo.


Subject(s)
Antigens, Surface/immunology , Forkhead Transcription Factors/immunology , Immune Tolerance , Interleukin-10/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Vacuolar Proton-Translocating ATPases/immunology , Animals , Antigens, Neoplasm , Antigens, Surface/metabolism , Forkhead Transcription Factors/biosynthesis , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , T-Lymphocytes, Regulatory/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
5.
FEBS Lett ; 581(5): 967-74, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17306257

ABSTRACT

The adapter 3BP2 is involved in leukocyte signaling downstream Src/Syk-kinases coupled immunoreceptors. Here, we show that 3BP2 directly interacts with the endocytic scaffold protein CIN85 and the actin-binding protein HIP-55. 3BP2 co-localized with CIN85 and HIP-55 in T cell rafts and at the T cell/APC synapse, an active zone of receptors and proteins recycling. A binding region of CIN85 SH3 domains on 3BP2 was mapped to a PVPTPR motif in the first proline-rich region of 3BP2, whereas the C-terminal SH3 domain of HIP-55 bound a more distal proline-rich domain of 3BP2. Together, our data suggest an unexpected role of 3BP2 in endocytic and cytoskeletal regulation through its interaction with CIN85 and HIP-55.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Microfilament Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Cell Line , Cytoskeleton/metabolism , Endocytosis , Humans , In Vitro Techniques , Jurkat Cells , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Models, Biological , Proline/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , T-Lymphocytes/metabolism , Two-Hybrid System Techniques , src Homology Domains/genetics
6.
Cell Signal ; 19(7): 1404-18, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17303381

ABSTRACT

Exogenous bacterial sphingomyelinase (SMase) and C6-Ceramides (C6-Cer) considerably lower buoyant cholesterol on sucrose density-gradient (at least 55% less cholesterol). In opposition, short C2-Cer fails to displace buoyant cholesterol. Note that neither SMase nor C6-Cer delocalize raft markers (Lck, LAT, CD55, and GM1). They are still anchored in ceramides-rich/cholesterol-poor domains, demonstrating that cholesterol is not necessary for their buoyancy. SMase-treated cells, i.e. cells exhibiting cholesterol-depleted rafts, optimally transmit CD3-induced phosphorylations (tyrosine, threonine, and serine). SMase, that extracts and partially displaces buoyant cholesterol, does not inhibit PLCgamma1-LAT interaction, Vav 1 phosphorylation, the actin polymerization, IL-2 and NF-kappaB (EMSA and luciferase assays) activation, and CD25 up-regulation (RT-PCR and cytometry) at all. Nevertheless, Ca(2+) influx and diacylglycerol (palmitoyl-DAG and arachidonoy-DAG) production are lowered. The drop of CD3-induced Ca(2+) influx is due to a strong plasma membrane depolarization because of Cer. The decreased DAG level is a consequence of the drop of intracellular Ca(2+) that is a cofactor for the PLCgamma1. In conclusion, our study challenges the real role of cholesterol-rich rafts in CD3/TCR signaling and suggests that other membrane domains than cholesterol-rich rafts can optimally transmit CD3/TCR signals.


Subject(s)
CD3 Complex/metabolism , Cholesterol/deficiency , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Receptors, Antigen, T-Cell/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Biomarkers/metabolism , Calcium Signaling , Cell Survival , Ceramides/metabolism , Cytoskeleton/metabolism , Cytosol/enzymology , Diglycerides/metabolism , Humans , Hydrolysis , Interleukin-2 Receptor alpha Subunit/metabolism , Jurkat Cells , MAP Kinase Kinase 1/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phospholipase C gamma/metabolism , Phosphotyrosine/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism
8.
Cell Signal ; 18(1): 105-22, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15925486

ABSTRACT

T lymphocytes contain two kinetic pools of cholesterol extractable with methyl-beta-cyclodextrin (m-beta-CD): a fast pool (31.5%, t1/2=17 s) and a slow pool (68.5%, t1/2=15 min). Purification of detergent-resistant membranes (DRMs) shows that the fast pool corresponds to buoyant cholesterol. Cholesterol extraction of the fast pool (i.e. cholesterol from rafts) still allows the buoyancy of signaling proteins and their phosphorylation under CD3 stimulation. Cholesterol depletion of the slow pool (i.e. cholesterol from membranes other than rafts) is accompanied by the extraction of the whole raft followed by the inhibition of CD3-induced tyrosine-phosphorylations. Cholesterol oxidase (COase) allows a specific oxidation of raft cholesterol into cholestenone. Cholestenone leaves the DRMs and accumulates as Triton X-100-soluble material. Specific cholesterol-rich raft disruption by COase does not inhibit the activation of either Jurkat cells or T CD4+ lymphocytes. Our study challenges the real role of cholesterol-rich rafts in CD3/TCR signaling and suggests that a cholesterol-poor subtype of rafts is involved in signal transmission via the TCR.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Cholesterol/physiology , Membrane Microdomains/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction/physiology , Adult , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholesterol/metabolism , Dose-Response Relationship, Drug , Humans , Jurkat Cells , Membrane Microdomains/drug effects , Oxidation-Reduction , Phospholipase C gamma/metabolism , Phosphorylation , Receptors, Antigen, T-Cell/drug effects , Reference Values , Signal Transduction/drug effects , Time Factors , Tyrosine/metabolism , beta-Cyclodextrins/pharmacology
9.
J Immunol ; 175(9): 5637-48, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16237053

ABSTRACT

The inhibition of human CD4+ T lymphocyte activation and proliferation by cholera toxin B-subunit (CTB) is a well-established phenomenon; nevertheless, the exact mechanism remained unclear. In the present study, we propose an explanation for the rCTB-induced inhibition of CD4+ T lymphocytes. rCTB specifically binds to GM1, a raft marker, and strongly modifies the lipid composition of rafts. First, rCTB inhibits sphingomyelin synthesis; second, it enhances phosphatidylcholine synthesis; and third, it activates a raft-resident neutral sphingomyelinase resembling to neutral sphingomyelinase type 1, thus generating a transient ceramide production. We demonstrated that these ceramides inhibit protein kinase Calpha phosphorylation and its translocation into the modified lipid rafts. Furthermore, we show that rCTB-induced ceramide production activate NF-kappaB. Combined all together: raft modification in terms of lipids, ceramide production, protein kinase Calpha inhibition, and NF-kappaB activation lead to CD4+ T cell inhibition.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cholera Toxin/pharmacology , Lymphocyte Activation/drug effects , Membrane Microdomains/enzymology , Sphingomyelin Phosphodiesterase/physiology , Acetylcysteine/pharmacology , Adult , CD4-Positive T-Lymphocytes/immunology , Ceramides/biosynthesis , Enzyme Activation/drug effects , G(M1) Ganglioside/biosynthesis , G(M1) Ganglioside/pharmacology , Glutathione/pharmacology , Humans , Ionomycin/pharmacology , NF-kappa B/metabolism , Phosphorylation , Protein Kinase C-alpha/metabolism , Protein Transport , Sphingomyelins/metabolism
10.
J Immunol ; 175(2): 1214-23, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16002725

ABSTRACT

The VLA-4 integrin supports static cell-cell, cell-matrix adhesion, and dynamic interactions with VCAM-1. Although functions for well-conserved beta(1) integrin cytoplasmic domains in regulating static cell adhesion has been established, the molecular basis for beta(1) integrin-dependent arrest on VCAM-1 under flow conditions remains poorly understood. We have transfected the beta(1) integrin-deficient A1 Jurkat T cell line with beta(1) cDNA constructs with deletions of the NPXY motifs and specific mutations of tyrosine residues. Deletion of either NPXY motif impaired static adhesion induced by CD2 or CD47 triggering or direct beta(1) integrin stimulation. In contrast, PMA-induced adhesion to VCAM-1 was unaffected by deletion of the NPIY motif and only slightly impaired by deletion of NPKY. Moreover, deletion of the NPIY motif resulted in enhanced rolling and reduced arrest on VCAM-1 under shear flow conditions. In contrast, deletion of the NPKY motif did not alter arrest under flow. Although tyrosine to phenylalanine substitutions within two NPXY motifs did not alter static adhesion to VCAM-1, these mutations enhanced arrest on VCAM-1 under flow conditions. Furthermore, although deletion of the C'-terminal 5 AA of the beta(1) cytoplasmic domain dramatically impaired activation-dependent static adhesion, it did not impair arrest on VCAM-1 under flow conditions. Thus, our results demonstrate distinct structural requirements for VLA-4 function under static and shear flow conditions. This may be relevant for VLA-4 activity regulation in different anatomic compartments, such as when circulating cells arrest on inflamed endothelium under shear flow and when resident cells in bone marrow interact with VCAM-1- positive stromal cells.


Subject(s)
Cell Movement , Cytoplasm/physiology , Integrin alpha4beta1/physiology , Integrin beta1/physiology , T-Lymphocyte Subsets/physiology , Vascular Cell Adhesion Molecule-1/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Migration Inhibition , Cell Movement/genetics , Cytoplasm/genetics , Fibronectins/metabolism , Humans , Integrin alpha4beta1/chemistry , Integrin alpha4beta1/metabolism , Integrin beta1/biosynthesis , Integrin beta1/chemistry , Integrin beta1/genetics , Jurkat Cells , Molecular Sequence Data , Peptide Fragments/genetics , Protein Structure, Tertiary/genetics , Sequence Deletion , T-Lymphocyte Subsets/metabolism , Tyrosine/genetics , Vascular Cell Adhesion Molecule-1/physiology
11.
J Exp Med ; 201(9): 1397-405, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15867092

ABSTRACT

Here, we report the isolation of a human multipotent adipose-derived stem (hMADS) cell population from adipose tissue of young donors. hMADS cells display normal karyotype; have active telomerase; proliferate >200 population doublings; and differentiate into adipocytes, osteoblasts, and myoblasts. Flow cytometry analysis indicates that hMADS cells are CD44+, CD49b+, CD105+, CD90+, CD13+, Stro-1(-), CD34-, CD15-, CD117-, Flk-1(-), gly-A(-), CD133-, HLA-DR(-), and HLA-I(low). Transplantation of hMADS cells into the mdx mouse, an animal model of Duchenne muscular dystrophy, results in substantial expression of human dystrophin in the injected tibialis anterior and the adjacent gastrocnemius muscle. Long-term engraftment of hMADS cells takes place in nonimmunocompromised animals. Based on the small amounts of an easily available tissue source, their strong capacity for expansion ex vivo, their multipotent differentiation, and their immune-privileged behavior, our results suggest that hMADS cells will be an important tool for muscle cell-mediated therapy.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation , Dystrophin/metabolism , Gene Expression Regulation , Immunocompetence/physiology , Multipotent Stem Cells/transplantation , Animals , Child , Child, Preschool , DNA Primers , Female , Flow Cytometry , Humans , Immunohistochemistry , Infant , Karyotyping , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
12.
J Infect ; 49(3): 216-21, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15337338

ABSTRACT

OBJECTIVE: Because the absence of immune restoration in HIV-infected patients efficiently treated by highly active antiretroviral therapy (HAART) may be due to excessive immune activation, we prospectively studied the effect of hydrocortisone on T-cell apoptosis in a cohort of patients with satisfactory virologic response. METHODS: Apoptosis of T-cell subsets including naïve CD45RA(+)CD4+ T-cells was determined at baseline and at months 1 and 3 after initiation of HAART. A satisfactory immune response was defined as an increase >100/microL CD4+ T-cells at month 3 compared to baseline. RESULTS: Twenty out of 63 patients showed undetectable viral load at month 3, among whom eight exhibited a satisfactory immune response. Down-regulation spontaneous CD4+T-cell apoptosis was significant in the group of patients with a satisfactory immune response compared to the other patients. However, hydrocortisone up-regulated apoptosis of naïve CD4+ CD45RA+ T-cells, specifically in group of patients with poor immune response, whatever the time point considered: percentage of apoptotic CD4 T-cells was 16+/-16% without hydrocortisone and 22+/-22% with hydrocortisone at month 1, and respectively, 10+/-9 and 17+/-15% at month 3 (P < 0.05) Hydrocortisone had no impact on CD8+ T-cell apoptosis, whatever the considered group. CONCLUSION: Our results suggest to not use steroid therapy as adjuvant immunotherapy in patients with less than optimal immunologic response to HAART.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Apoptosis/drug effects , CD4 Antigens/drug effects , HIV Infections/drug therapy , Hydrocortisone/therapeutic use , T-Lymphocyte Subsets/drug effects , Adult , Anti-Inflammatory Agents/administration & dosage , CD4 Antigens/immunology , Chemotherapy, Adjuvant , Female , HIV Infections/immunology , Humans , Hydrocortisone/administration & dosage , Male , Middle Aged , Prospective Studies , T-Lymphocyte Subsets/immunology , Viral Load
13.
J Immunol ; 172(3): 1397-406, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734715

ABSTRACT

We have previously shown that laminin-5 is expressed in the human thymic medulla, in which mature thymocytes are located. We now report that laminin-5 promotes migration of mature medullary thymocytes, whereas it has no effect on cortical immature thymocytes. Migration was inhibited by blocking mAbs directed against laminin-5 integrin receptors and by inhibitors of metalloproteinases. Interactions of thymocytes with laminin-5 induced a strong up-regulation of active metalloproteinase-14. However, we found that thymocytes did not cleave the laminin-5 gamma(2) chain, suggesting that they do not use the same pathway as epithelial cells to migrate on laminin-5. Interactions of thymocytes with laminin-5 also induced the release of a soluble fragment of CD44 cell surface molecule. Moreover, CD44-rich supernatants induced thymocyte migration in contrast with supernatants depleted in CD44 by immunoadsorption. CD44 cleavage was recently reported to be due to metalloproteinase-14 activation and led to increased migration in cancer cells. Thus, in this study, we show that laminin-5 promotes human mature thymocyte migration in vitro via a multimolecular mechanism involving laminin-5 integrin receptors, metalloproteinase-14 and CD44. These data suggest that, in vivo, laminin-5 may function in the migration of mature thymocytes within the medulla and be part of the thymic emigration process.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Movement/immunology , Hyaluronan Receptors/metabolism , Metalloendopeptidases/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Cell Communication/immunology , Cell Differentiation/immunology , Cells, Cultured , Enzyme Activation/immunology , Humans , Hyaluronan Receptors/physiology , Hydrolysis , Immunophenotyping , Infant , Integrin alpha3beta1/physiology , Integrin alpha6beta4/physiology , Laminin/metabolism , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/physiology , Peptide Fragments/metabolism , Protein Processing, Post-Translational/immunology , Protein Subunits/metabolism , Solubility , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Thymus Gland/enzymology , Thymus Gland/immunology , Kalinin
14.
J Immunol ; 171(10): 5018-26, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14607898

ABSTRACT

There is now compelling evidence that CD4(+)CD25(+) T cells play a major role in the maintenance of tolerance. Besides CD4(+)CD25(+) T cells, different populations of regulatory CD4(+) T cells secreting high amounts of IL-10 (T regulatory type 1 (Tr1)) or TGF-beta (Th3) have also been described in in vivo models. In the lymphocyte transfer model of inflammatory bowel disease, we show here that the control of inflammation during the first weeks is not due to a complete inhibition of differentiation of aggressive proinflammatory T cells, but is the result of a balance between proinflammatory and Tr cells. We also show that in the first weeks continuous IL-10 secretion was required to actively control inflammation. Indeed, treatment with anti-IL-10R Abs 3 wk after the start of the experiment completely reversed the protective effect of Tr cells. IL-10 secretion and control of inflammation could be provided by late injection of Tr1 cells that efficiently cure ongoing inflammatory responses in two different models of inflammation. In contrast, inflammation was not controlled when high numbers of CD4(+)CD45RB(low) or CD4(+)CD25(+) T cells were injected as early as 1 wk after the start of the experiment. These results confirm in vitro studies showing that CD4(+)CD45RB(low) do not contain high IL-10-producing cells and suggest that CD4(+)CD45RB(low) Tr cells maintain tolerance in vivo, in part indirectly, through the differentiation of IL-10-secreting Tr1 cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/prevention & control , Receptors, Interleukin-2/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Clone Cells , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Dermatitis, Contact/prevention & control , Disease Models, Animal , Female , Inflammatory Bowel Diseases/pathology , Interleukin-10/physiology , Mice , Mice, Inbred BALB C , Mice, SCID , Skin/immunology , Skin/pathology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Th1 Cells/transplantation , Th2 Cells/transplantation
15.
J Antimicrob Chemother ; 52(2): 269-75, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12837745

ABSTRACT

Multiple failures of antiretroviral treatments, as a result of multidrug-resistant virus, have led to a proposal for structured therapeutic interruptions (STI). However, a significant decrease in CD4+ T cells may occur. The aim of our study was to determine the kinetics of T cell subpopulation changes, T cell apoptosis and peripheral blood mononuclear cell proliferation after STI. The impact of resistance mutation disappearance on T cell apoptosis was also studied. Ten patients were enrolled prospectively, and blood sampling was performed at weeks 0, 2, 4, 6, 8 and 12. The mean increase in viral load was 1.3 log(10) copies/ml, ranging from 0.1 to 3.2. CD4+ T cell count decreased to a mean of 80 cells/mm(3) from baseline to week 12. In the same period, CD8+ T cells decreased to a mean of 139 cells/mm(3). A significant increase in both T cell apoptosis and proliferation of mononuclear cells was observed. However, proliferation was an early and brief event. The increase in CD4+ T cell apoptosis was obvious in patients exhibiting complete reversion of resistance mutations to antiviral drugs. Our results suggest that during STI, apoptosis is an overwhelming phenomenon compared with proliferation, and may explain the limited immunological impact of this therapeutic option.


Subject(s)
Antiretroviral Therapy, Highly Active/adverse effects , Apoptosis/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Apoptosis/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Division/drug effects , Cell Division/physiology , Drug Administration Schedule , Humans , Longitudinal Studies , Prospective Studies , T-Lymphocytes/cytology
16.
Genetics ; 164(3): 1015-25, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12871911

ABSTRACT

Multicellular organisms grow through both proliferation and growth of their individual cells. We have conducted a P-element-based misexpression screen for genes whose upregulation alters wing disc growth during development. One particular group of four P elements, all inserted at cytological location 61C7-8, exhibited specific overgrowth upon misexpression in proliferating imaginal tissues. Clonal analysis revealed that upon misexpression, cell number was increased but cell size was not affected, indicating that cell growth and proliferation were induced in a coordinate manner. Loss of function at the locus produced small flies with reduced cell number, consistent with the presence of a gene encoding a positive growth regulator. We characterized a new transcription unit initiating in a region adjacent to the P insertions, which generated a complex series of polyadenylated transcripts. Although these RNAs were induced in response to misexpression, none was sufficient by itself to recapitulate overgrowth when overexpressed. This suggested either that a particular combination of these transcripts was necessary or that other sequences are involved.


Subject(s)
Cell Division/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression , Wings, Animal/growth & development , Animals , Blotting, Northern , Body Weights and Measures , Chromosome Mapping , Crosses, Genetic , DNA Primers , Embryo, Nonmammalian/embryology , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Wings, Animal/embryology
17.
Immunity ; 18(5): 605-17, 2003 May.
Article in English | MEDLINE | ID: mdl-12753738

ABSTRACT

Active suppression is mediated by a subpopulation of CD4(+) T cells that prevents autoimmunity. However, the mechanisms involved in their differentiation in vivo are currently under intensive research. Here we show that in vitro culture of bone marrow cells in the presence of IL-10 induces the differentiation of a distinct subset of dendritic cells with a specific expression of CD45RB. These CD11c(low)CD45RB(high) DCs are present in the spleen and lymph nodes of normal mice and are significantly enriched in the spleen of IL-10 Tg mice. These natural or in vitro-derived DCs display plasmacytoid morphology and an immature-like phenotype, and secrete high levels of IL-10 after activation. OVA peptide-pulsed CD11c(low)CD45RB(high) DCs specifically induce tolerance through the differentiation of Tr1 cells in vitro and in vivo. Our findings identify a natural DC subset that induces the differentiation of Tr1 cells and suggest their therapeutic use.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Interleukin-10/immunology , Animals , Interferon-alpha/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
18.
Biochem J ; 363(Pt 3): 645-55, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11964165

ABSTRACT

Cell membranes contain sphingolipids and cholesterol, which cluster together in distinct domains called rafts. The outer-membrane leaflet of these peculiar membrane domains contains glycosylphosphatidylinositol-anchored proteins, while the inner leaflet contains proteins implicated in signalling, such as the acylated protein kinase p56(lck) and the palmitoylated adaptator LAT (linker for activation of T-cells). We present here an approach to study the lipid composition of rafts and its change upon T-cell activation. Our method is based on metabolic labelling of Jurkat T-cells with different precursors of glycerophospholipid synthesis, including glycerol and fatty acids with different lengths and degrees of saturation as well as phospholipid polar head groups. The results obtained indicate that lipid rafts isolated by the use of sucrose density-gradient centrifugation after Triton X-100 extraction in the cold, besides sphingolipids and cholesterol, contain unambiguously all classes of glycerophospholipids: phosphatidylserine, phosphatidylinositol, phosphatidylethanolamine and phosphatidylcholine. Fatty acid labelling shows that lipid rafts are labelled preferentially with saturated fatty acids while the rest of the plasma membrane incorporates mostly long-chained polyunsaturated fatty acids. To see whether the raft composition as measured by metabolic labelling of phospholipids is involved in T-cell activation, we investigated the production of sn-1,2-diacylglycerol (DAG) in CD3-activated cells. DAG production occurs within rafts, confirming previous demonstration of protein kinase C translocation into membrane microdomains. Our data demonstrate that raft disorganization by methyl-beta-cyclodextrin impairs both CD3-induced DAG production and changes in cytosolic Ca(2+) concentration. These lines of evidence support the conclusion that the major events in T-cell activation occur within or due to lipid rafts.


Subject(s)
CD3 Complex/physiology , Signal Transduction , beta-Cyclodextrins , Arachidonic Acid/metabolism , CD3 Complex/metabolism , Calcium/metabolism , Cell Membrane/ultrastructure , Cholesterol/metabolism , Cyclodextrins/pharmacology , Diglycerides/biosynthesis , Glycerophospholipids/metabolism , Humans , Jurkat Cells , Membrane Lipids/metabolism , Palmitic Acid/metabolism , Protein Kinase C/metabolism
19.
J Infect Dis ; 185(4): 463-70, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11865398

ABSTRACT

This prospective study investigated the contributions of apoptosis and proliferation of CD4(+) T cells obtained by the introduction of a new antiretroviral treatment for human immunodeficiency virus infection. Virus load; T cell counts; apoptosis of T cell subsets, including naive cells; and proliferation were determined from treatment initiation to the third month in a cohort of patients. An increase in CD4(+) T cell count > or = 100 cells/microL over baseline was considered to be a satisfactory immune reconstitution. Sixty-nine patients completed the protocol, 22 of whom met our definition of a satisfactory immune reconstitution, showing a significantly more pronounced reduction in spontaneous CD4(+) T cell apoptosis at month 1 as well as month 3, compared with the other patients. In contrast, neither Fas-induced apoptosis down-regulation nor Fas-induced increased proliferation capacity was associated with a satisfactory immune reconstitution. Down-regulation of CD4(+) T cell apoptosis by antiretroviral treatment is the main mechanism associated with early CD4(+) T cell increase.


Subject(s)
Anti-HIV Agents/therapeutic use , Apoptosis/drug effects , CD4 Lymphocyte Count , HIV Infections/drug therapy , Adult , Aged , Anti-HIV Agents/administration & dosage , Cell Division/drug effects , Down-Regulation , Drug Therapy, Combination , Female , HIV Infections/immunology , HIV Infections/pathology , Humans , Lymphocyte Activation/drug effects , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...