Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 9(9): e1003599, 2013.
Article in English | MEDLINE | ID: mdl-24086130

ABSTRACT

The interplay between autophagy and intracellular pathogens is intricate as autophagy is an essential cellular response to fight against infections, whereas numerous microbes have developed strategies to escape this process or even exploit it to their own benefit. The fine tuned timing and/or selective molecular pathways involved in the induction of autophagy upon infections could be the cornerstone allowing cells to either control intracellular pathogens, or be invaded by them. We report here that measles virus infection induces successive autophagy signallings in permissive cells, via distinct and uncoupled molecular pathways. Immediately upon infection, attenuated measles virus induces a first transient wave of autophagy, via a pathway involving its cellular receptor CD46 and the scaffold protein GOPC. Soon after infection, a new autophagy signalling is initiated which requires viral replication and the expression of the non-structural measles virus protein C. Strikingly, this second autophagy signalling can be sustained overtime within infected cells, independently of the expression of C, but via a third autophagy input resulting from cell-cell fusion and the formation of syncytia. Whereas this sustained autophagy signalling leads to the autophagy degradation of cellular contents, viral proteins escape from degradation. Furthermore, this autophagy flux is ultimately exploited by measles virus to limit the death of infected cells and to improve viral particle formation. Whereas CD150 dependent virulent strains of measles virus are unable to induce the early CD46/GOPC dependent autophagy wave, they induce and exploit the late and sustained autophagy. Overall, our work describes distinct molecular pathways for an induction of self-beneficial sustained autophagy by measles virus.


Subject(s)
Measles virus/metabolism , Measles virus/pathogenicity , Measles/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Antigens, CD/genetics , Antigens, CD/metabolism , Autophagy , Carrier Proteins/genetics , Carrier Proteins/metabolism , Giant Cells/metabolism , Giant Cells/pathology , Giant Cells/virology , Golgi Matrix Proteins , HeLa Cells , Humans , Measles/genetics , Measles/pathology , Measles virus/genetics , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
2.
PLoS Pathog ; 7(12): e1002422, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174682

ABSTRACT

Autophagy is a conserved degradative pathway used as a host defense mechanism against intracellular pathogens. However, several viruses can evade or subvert autophagy to insure their own replication. Nevertheless, the molecular details of viral interaction with autophagy remain largely unknown. We have determined the ability of 83 proteins of several families of RNA viruses (Paramyxoviridae, Flaviviridae, Orthomyxoviridae, Retroviridae and Togaviridae), to interact with 44 human autophagy-associated proteins using yeast two-hybrid and bioinformatic analysis. We found that the autophagy network is highly targeted by RNA viruses. Although central to autophagy, targeted proteins have also a high number of connections with proteins of other cellular functions. Interestingly, immunity-associated GTPase family M (IRGM), the most targeted protein, was found to interact with the autophagy-associated proteins ATG5, ATG10, MAP1CL3C and SH3GLB1. Strikingly, reduction of IRGM expression using small interfering RNA impairs both Measles virus (MeV), Hepatitis C virus (HCV) and human immunodeficiency virus-1 (HIV-1)-induced autophagy and viral particle production. Moreover we found that the expression of IRGM-interacting MeV-C, HCV-NS3 or HIV-NEF proteins per se is sufficient to induce autophagy, through an IRGM dependent pathway. Our work reveals an unexpected role of IRGM in virus-induced autophagy and suggests that several different families of RNA viruses may use common strategies to manipulate autophagy to improve viral infectivity.


Subject(s)
Autophagy/physiology , GTP-Binding Proteins/metabolism , RNA Virus Infections/metabolism , RNA Virus Infections/transmission , RNA Viruses/metabolism , Base Sequence , Blotting, Western , Computational Biology , GTP-Binding Proteins/genetics , HeLa Cells , Humans , Microscopy, Confocal , Molecular Sequence Data , Open Reading Frames/genetics , RNA Virus Infections/genetics , RNA Viruses/genetics , RNA, Small Interfering , Transfection , Two-Hybrid System Techniques , Viral Proteins/metabolism
3.
Cell Host Microbe ; 6(4): 354-66, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19837375

ABSTRACT

Autophagy is a highly regulated self-degradative mechanism required at a basal level for intracellular clearance and recycling of cytoplasmic contents. Upon intracellular pathogen invasion, autophagy can be induced as an innate immune mechanism to control infection. Nevertheless, pathogens have developed strategies to avoid or hijack autophagy for their own benefit. The molecular pathways inducing autophagy in response to infection remain poorly documented. We report here that the engagement of CD46, a ubiquitous human surface receptor able to bind several different pathogens, is sufficient to induce autophagy. CD46-Cyt-1, one of the two C-terminal splice variants of CD46, is linked to the autophagosome formation complex VPS34/Beclin1 via its interaction with the scaffold protein GOPC. Measles virus and group A Streptococcus, two CD46-binding pathogens, induce autophagy through a CD46-Cyt-1/GOPC pathway. Thus, upon microorganism recognition, a cell surface pathogen receptor can directly trigger autophagy, a critical step to control infection.


Subject(s)
Autophagy , Measles virus/immunology , Membrane Cofactor Protein/immunology , Streptococcus pyogenes/immunology , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Carrier Proteins/metabolism , Golgi Matrix Proteins , Humans , Membrane Proteins/metabolism , Membrane Transport Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Interaction Mapping
4.
Nat Med ; 14(1): 81-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157139

ABSTRACT

IL-17A is a T cell-specific cytokine that is involved in chronic inflammations, such as Mycobacterium infection, Crohn's disease, rheumatoid arthritis and multiple sclerosis. Mouse models have explained the molecular basis of IL-17A production and have shown that IL-17A has a positive effect not only on granuloma formation and neurodegeneration through unknown mechanisms, but also on bone resorption through Receptor activator of NF-kappaB ligand (RANKL) induction in osteoblasts. Langerhans cell histiocytosis (LCH) is a rare disease of unknown etiology, lacking an animal model, that cumulates symptoms that are found separately in various IL-17A-related diseases, such as aggressive chronic granuloma formation, bone resorption and soft tissue lesions with occasional neurodegeneration. We examined IL-17A in the context of LCH and found that there were high serum levels of IL-17A during active LCH and unexpected IL-17A synthesis by dendritic cells (DCs), the major cell type in LCH lesions. We also found an IL-17A-dependent pathway for DC fusion, which was highly potentiated by IFN-gamma and led to giant cells expressing three major tissue-destructive enzymes: tartrate resistant acidic phosphatase and matrix metalloproteinases 9 and 12. IFN-gamma expression has been previously documented in LCH and observed in IL-17A-related diseases. Notably, serum IL-17A-dependent fusion activity correlates with LCH activity. Thus, IL-17A and IL-17A-stimulated DCs represent targets that may have clinical value in the treatment of LCH and other IL-17A-related inflammatory disorders.


Subject(s)
Dendritic Cells/metabolism , Histiocytosis, Langerhans-Cell/pathology , Interleukin-17/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Cell Fusion , Humans , Inflammation , Interferon-gamma/metabolism , Lymphocyte Activation , Lymphocytes/metabolism , Mice , Monocytes/metabolism , Mycobacterium/metabolism , Oligonucleotide Array Sequence Analysis
5.
Eur J Immunol ; 37(3): 747-57, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17304626

ABSTRACT

Dendritic cells (DC) are the mononuclear cells that initiate adaptive immune responses. Osteoclasts (OC) are the multinucleated giant cells that resorb bone. As previously described for human conventional DC (cDC), we demonstrate that murine cDC, either in vitro generated from Fms-like tyrosine kinase 3 (Flt3)+ bone marrow progenitors or ex vivo purified from spleen, are able to develop into OC in response to M-CSF and receptor activator of NF-kappaB ligand (RANKL) in vitro. This transdifferentiation is driven by the immune environment that controls cDC maturation, cell fusion, tartrate-resistant acid phosphatase (TRAP) and bone resorption activities. Only immature cDC have the capacity to become OC since mature cDC or plasmacytoid DC do not. Additions of the pro-inflammatory cytokines, such as IL-1beta and TNF-alpha, or human rheumatoid synovial fluid, increase murine cDC transdifferentiation into OC, whereas IFN-alpha inhibits it. The adaptive cytokine, IFN-gamma, inhibits cDC fusion while IL-4 increases it. IL-2, IFN-gamma and IL-4 inhibit TRAP and bone resorption activities contrary to IL-10, which enhances both activities. A putative new "immune multinucleated giant cell" unable to resorb bone, which is formed owing to IL-4, is underlined. The future analysis of cDC transdifferentiation into OC in murine models of inflammatory arthritis will give us the quantitative importance of this phenomenon in vivo.


Subject(s)
Cell Differentiation/immunology , Cytokines/physiology , Dendritic Cells/cytology , Growth Inhibitors/physiology , Osteoclasts/cytology , Animals , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Immunity, Active , Immunity, Innate , Mice , Mice, Inbred C57BL , Osteoclasts/immunology , Osteoclasts/metabolism
6.
J Immunol ; 177(8): 4957-61, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015676

ABSTRACT

T regulatory cell 1 (Tr1) are low proliferating peripherally induced suppressive T cells. Engaging CD3 and CD46 on human CD4+ T cells induces a Tr1-like phenotype. In this study, we report that human Tr1-like cells do not sustain proliferation over time. The weak proliferation of these cells results first from their inability to sustain expression of various cell cycle-associated proteins, to efficiently degrade the inhibitor of cell cycle progression p27/Kip1 and, as a consequence, in their accumulation in the G0-G1 phase. Also, the reduced proliferation of Tr1-like cells results from their increased sensitivity to death as they divide, through a mechanism that is neither Fas-mediated nor Bcl2/Bcl-xL related. Both properties, impaired cell cycle and death sensitivity, are explained by a specific defective activation of Akt that impairs the expression of Survivin. Thus, our results show that CD3/CD46-induced Tr1-like cells die through a process of abortive proliferation.


Subject(s)
Cell Proliferation , Membrane Cofactor Protein/physiology , Microtubule-Associated Proteins/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , T-Lymphocytes, Regulatory/cytology , CD3 Complex/physiology , Cell Cycle , Cell Death , Cyclin-Dependent Kinase Inhibitor p27 , Humans , Inhibitor of Apoptosis Proteins , Intracellular Signaling Peptides and Proteins/metabolism , Survivin
7.
J Immunol ; 172(9): 5405-14, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15100281

ABSTRACT

High doses of Ag can paradoxically suppress immune responses in vivo. This Ag-specific unresponsiveness (termed high dose tolerance) involves extrathymic mechanisms in mature T lymphocytes. To investigate these mechanisms, we used the in vitro model of PBL activated with anti-CD3 or PHA. In these conditions, increasing mitogen concentrations resulted in a reduction of the proliferative response, associated with an increased percentage of apoptotic cells. Apoptosis did not require prior exposure to IL-2, it was not the consequence of CD178/CD95 or TNF/TNFR interactions, and was therefore clearly distinct from activation-induced cell death. Although the pan-caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (zVAD-fmk) decreased DNA fragmentation, cytochrome c release and caspase-9 and caspase-3 activation were not implicated, suggesting that this apoptosis did not primarily involve the intrinsic mitochondrial pathway. E64d, a cysteine protease inhibitor, as well as specific inhibitors of cathepsin B and cathepsin L conferred protection. We further demonstrated that cathepsin B and cathepsin L were released from the lysosomes and catalytically active in the cytosol. Release of cathepsin B and cathepsin L was the consequence of lysosomal membrane permeabilization without complete disruption of the cytosol-lysosome pH gradient. These results demonstrate a role for cathepsins in supraoptimal activation-induced apoptosis in vitro and suggest their possible participation in high dose tolerance in vivo.


Subject(s)
Apoptosis/immunology , Cathepsin B/physiology , Cathepsins/physiology , Lymphocyte Activation/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Amino Acid Chloromethyl Ketones/pharmacology , CD28 Antigens/pharmacology , Caspase Inhibitors , Catalysis , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cathepsin L , Cathepsins/antagonists & inhibitors , Cathepsins/metabolism , Cell Death/immunology , Cell Differentiation/immunology , Cells, Cultured , Cysteine Endopeptidases , Cysteine Proteinase Inhibitors/pharmacology , Cytochromes c/metabolism , Cytosol/enzymology , Cytosol/immunology , Cytosol/metabolism , DNA Fragmentation/drug effects , DNA Fragmentation/immunology , Dose-Response Relationship, Immunologic , G1 Phase/immunology , Humans , Intracellular Membranes/enzymology , Intracellular Membranes/immunology , Lysosomes/enzymology , Muromonab-CD3/pharmacology , Permeability , S Phase/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology
8.
Blood ; 102(10): 3719-26, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12893746

ABSTRACT

Antithymocyte globulins (ATGs), the immunoglobulin G (IgG) fraction of sera from rabbits or horses immunized with human thymocytes or T-cell lines, are used in conditioning regimens for bone marrow transplantation, in the treatment of acute graft-versus-host disease, in the prevention or treatment of acute rejection in organ transplantation, and in severe bone marrow aplasia. In nonhuman primates, ATGs induce rapid, dose-dependent, T-cell depletion in peripheral lymphoid tissues, where apoptotic cells can be demonstrated in T-cell zones. We show here that increasing ATG concentrations in vitro resulted in reduced lymphocyte proliferative responses, associated with a rapid increase in the percentage of apoptotic cells. Apoptosis did not require prior exposure to interleukin-2, nor did it result in CD178/CD95 or tumor necrosis factor/tumor necrosis factor receptor (TNF/TNF-R) interactions; it was therefore clearly different from activation-induced cell death. Cytochrome c release, caspase-9, and caspase-3 activation were not implicated, excluding a direct involvement of the intrinsic mitochondrial pathway. The cysteine protease inhibitor E64d and cathepsin-B-specific inhibitors conferred significant protection, whereas apoptosis was associated with the release of active cathepsin B into the cytosol. These data demonstrate a role for cathepsin B in T-cell apoptosis induced by ATGs at concentrations achieved during clinical use.


Subject(s)
Antilymphocyte Serum/pharmacology , Apoptosis , Cathepsin B/physiology , Lymphocyte Depletion/methods , Antilymphocyte Serum/administration & dosage , Caspases , Cathepsin B/metabolism , Cytochromes c , Cytosol , Dose-Response Relationship, Drug , Humans , Lymphocyte Activation/drug effects , Lysosomes , T-Lymphocytes/cytology
9.
J Immunol ; 170(10): 4986-95, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12734342

ABSTRACT

Purine and pyrimidine nucleotides play critical roles in DNA and RNA synthesis as well as in membrane lipid biosynthesis and protein glycosylation. They are necessary for the development and survival of mature T lymphocytes. Activation of T lymphocytes is associated with an increase of purine and pyrimidine pools. However, the question of how purine vs pyrimidine nucleotides regulate proliferation, cell cycle, and survival of primary T lymphocytes following activation has not yet been specifically addressed. This was investigated in the present study by using well-known purine (mycophenolic acid, 6-mercaptopurine) and pyrimidine (methotrexate, 5-fluorouracil) inhibitors, which are used in neoplastic diseases or as immunosuppressive agents. The effect of these inhibitors was analyzed according to their time of addition with respect to the initiation of mitogenic activation. We showed that synthesis of both purine and pyrimidine nucleotides is required for T cell proliferation. However, purine and pyrimidine nucleotides differentially regulate the cell cycle since purines control both G(1) to S phase transition and progression through the S phase, whereas pyrimidines only control progression from early to intermediate S phase. Furthermore, inhibition of pyrimidine synthesis induces apoptosis whatever the time of inhibitor addition whereas inhibition of purine nucleotides induces apoptosis only when applied to already cycling T cells, suggesting that both purine and pyrimidine nucleotides are required for survival of cells committed into S phase. These findings reveal a hitherto unknown role of purine and pyrimidine de novo synthesis in regulating cell cycle progression and maintaining survival of activated T lymphocytes.


Subject(s)
Cell Cycle/immunology , Purine Nucleotides/pharmacology , Pyrimidine Nucleotides/pharmacology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Apoptosis/drug effects , Apoptosis/immunology , Cell Aggregation/drug effects , Cell Aggregation/immunology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Division/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , G1 Phase/drug effects , G1 Phase/immunology , Growth Inhibitors/pharmacology , Humans , Kinetics , Lymphocyte Activation/drug effects , Lymphocyte Count , Nucleic Acid Synthesis Inhibitors/pharmacology , Purine Nucleotides/antagonists & inhibitors , Purine Nucleotides/biosynthesis , Pyrimidine Nucleotides/antagonists & inhibitors , Pyrimidine Nucleotides/biosynthesis , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/immunology , S Phase/drug effects , S Phase/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
10.
Transplantation ; 75(5): 657-62, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12640305

ABSTRACT

BACKGROUND: Polyclonal antithymocyte globulins (ATG) induce T-cell depletion and functional impairment of nondeleted lymphocytes. Interference of ATG with the main leukocyte surface molecules involved in cellular adhesion and leukocyte-endothelium interaction was investigated in the present study. METHODS: In three rabbit ATG, the authors measured antibodies to integrins, beta2-integrin ligands, and chemokine receptors by flow cytometry; chemotactic responses; and down-modulation of cell surface expression on lymphocytes, monocytes, and neutrophils. RESULTS: Antibodies to CD11a/CD18 (leukocyte function-associated antigen-1 [LFA-1]) present in ATG induced a dose-dependent down-modulation of cell surface expression of this beta2 integrin on lymphocytes, monocytes, and neutrophils. In contrast, anti-LFA-1 monoclonal antibodies did not induce LFA-1 modulation unless cross-linked by a second antibody. ATG also contained functional antibodies to the beta1 integrin CD49d/CD29 (VLA-4), the alpha4beta7 integrin, CD50, CD54, and CD102 but not to CD62L. ATG were shown to bind to CXCR4 and CCR7 on lymphocytes, CXCR4, and CCR5 on monocytes; to down-modulate cell surface expression of CCR7; and to decrease monocyte chemotactic response to CCL5 (RANTES) and lymphocyte chemotactic response to CCL19 (MIP-3beta). CONCLUSION: These results show that ATG may interfere with leukocyte responses to chemotactic signals but mostly inhibit the expression of integrins required for firm cellular adhesion. The latter property of inhibition is not shared by monoclonal antibodies, and it may contribute to decreasing graft cellular infiltration during acute rejection and possibly after postischemic reperfusion.


Subject(s)
Antibodies/analysis , Antilymphocyte Serum/immunology , Cell Adhesion Molecules/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Antigens, CD/immunology , Down-Regulation/drug effects , Humans , Integrins/immunology , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Rabbits , Receptors, Chemokine/immunology , Selectins/immunology
11.
J Immunol ; 169(9): 4805-10, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391190

ABSTRACT

Exposure of phosphatidylserine (PS) on the outer leaflet of the plasma membrane is a key feature of apoptosis. As the signals underlying these phenomena are unknown, it is generally assumed that PS exposure is a consequence of caspase activation, another hallmark of apoptosis. In this study we investigated the role of caspases in PS externalization during apoptosis of activated PBL triggered by drugs (etoposide, staurosporine), CD95 engagement, or IL-2 withdrawal. Anti-CD95 mAb induces a rapid activation of caspases, followed by PS exposure and mitochondrial transmembrane potential (DeltaPsim) disruption. In contrast, etoposide (ETO), staurosporine (STS), or IL-2 withdrawal triggers concomitant caspase activation, PS exposure, and DeltaPsim disruption. Such kinetics suggest that PS exposure could be independent of caspase activation. As expected, in activated PBL treated by anti-CD95 mAb, the pan-caspase inhibitor Cbz-Val-Ala-Asp(OMe)-fluoromethylketone and the caspase-8 inhibitor Cbz-Leu-Glu-Thr-Asp(OMe)-fluoromethylketone, but not the caspase-9 inhibitor Cbz-Leu-Glu-His-Asp(OMe)-fluoromethylketone, inhibit PS externalization and DeltaPsim disruption. Surprisingly, during apoptosis induced by ETO, STS, or IL-2 withdrawal, none of those caspase inhibitors prevents PS externalization or DeltaPsim disruption, whereas they all inhibit DNA fragmentation as well as the morphological features of nuclear apoptosis. In Jurkat and H9 T cell lines, as opposed to activated PBL, PS exposure is inhibited by Cbz-Val-Ala-Asp(OMe)-fluoromethylketone during apoptosis induced by CD95 engagement, ETO, or STS. Thus, caspase-independent PS exposure occurs in primary T cells during apoptosis induced by stimuli that do not trigger death receptors.


Subject(s)
Apoptosis/immunology , Caspases/physiology , Phosphatidylserines/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Caspase 3 , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/immunology , Cells, Cultured , DNA Fragmentation/drug effects , DNA Fragmentation/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Etoposide/pharmacology , Humans , Interleukin-2/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/immunology , Intracellular Membranes/metabolism , Jurkat Cells , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/immunology , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/metabolism , Permeability/drug effects , Staurosporine/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Tumor Cells, Cultured
12.
J Immunol ; 169(5): 2747-55, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12193749

ABSTRACT

Mycophenolic acid (MPA), the active metabolite of the immunosuppressive drug mycophenolate mofetil, is a selective inhibitor of inosine 5'-monophosphate dehydrogenase type II, a de novo purine nucleotide synthesis enzyme expressed in T and B lymphocytes and up-regulated upon cell activation. In this study, we report that the blockade of guanosine nucleotide synthesis by MPA inhibits mitogen-induced proliferation of PBL, an effect fully reversed by addition of guanosine and shared with mizoribine, another inhibitor of inosine 5'-monophosphate dehydrogenase. Because MPA does not inhibit early TCR-mediated activation events, such as CD25 expression and IL-2 synthesis, we investigated how it interferes with cytokine-dependent proliferation and survival. In activated lymphoblasts that are dependent on IL-2 or IL-15 for their proliferation, MPA does not impair signaling events such as of the extracellular signal-regulated kinase 2 and Stat5 phosphorylation, but inhibits down-regulation of the cyclin-dependent kinase inhibitor p27(Kip1). Therefore, in activated lymphoblasts, MPA specifically interferes with cytokine-dependent signals that control cell cycle and blocks activated T cells in the mid-G(1) phase of the cell cycle. Although it blocks IL-2-mediated proliferation, MPA does not inhibit cell survival and Bcl-x(L) up-regulation by IL-2 or other cytokines whose receptors share the common gamma-chain (CD132). Finally, MPA does not interfere with IL-2-dependent acquisition of susceptibility to CD95-mediated apoptosis and degradation of cellular FLIP. Therefore, MPA has unique functional properties not shared by other immunosuppressive drugs interfering with IL-2R signaling events such as rapamycin and CD25 mAbs.


Subject(s)
Apoptosis/immunology , Immunosuppressive Agents/pharmacology , Interleukin-2/antagonists & inhibitors , Interleukin-2/physiology , Lymphocyte Activation/drug effects , Mycophenolic Acid/pharmacology , T-Lymphocytes/immunology , Apoptosis/drug effects , Cell Division/drug effects , Cell Division/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Growth Inhibitors/pharmacology , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/biosynthesis , Immunization , Interleukin-15/antagonists & inhibitors , Interleukin-15/physiology , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , fas Receptor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...