Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Publication year range
1.
Nat Rev Immunol ; 1(1): 11-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11905810

ABSTRACT

In the past decade, advances in genetic engineering and mouse knockout technology have transformed our understanding of the immune system. In particular, new perspectives on T-cell development, co-stimulation and activation have emerged from the study of single and multiple gene-knockout animals, as well as from conditional knockout and 'knock-in' mutants. Analysis of these animals has clarified important intracellular signalling pathways and has shed light on the regulatory mechanisms that govern normal immune responses and autoimmunity.


Subject(s)
Mice, Knockout/immunology , Animals , Gene Targeting , Genetic Engineering , Lymphocyte Activation , Mice , Mice, Knockout/genetics , Models, Immunological , Signal Transduction , T-Lymphocytes/immunology
2.
Acta Physiol (Oxf) ; 240(8): e14160, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38747650

ABSTRACT

AIM: Striatin (Strn) is a scaffold protein expressed in cardiomyocytes (CMs) and alteration of its expression are described in various cardiac diseases. However, the alteration underlying its pathogenicity have been poorly investigated. METHODS: We studied the role(s) of cardiac Strn gene (STRN) by comparing the functional properties of CMs, generated from Strn-KO and isogenic WT mouse embryonic stem cell lines. RESULTS: The spontaneous beating rate of Strn-KO CMs was faster than WT cells, and this correlated with a larger fast INa conductance and no changes in If. Paced (2-8 Hz) Strn-KO CMs showed prolonged action potential (AP) duration in comparison with WT CMs and this was not associated with changes in ICaL and IKr. Motion video tracking analysis highlighted an altered contraction in Strn-KO CMs; this was associated with a global increase in intracellular Ca2+, caused by an enhanced late Na+ current density (INaL) and a reduced Na+/Ca2+ exchanger (NCX) activity and expression. Immunofluorescence analysis confirmed the higher Na+ channel expression and a more dynamic microtubule network in Strn-KO CMs than in WT. Indeed, incubation of Strn-KO CMs with the microtubule stabilizer taxol, induced a rescue (downregulation) of INa conductance toward WT levels. CONCLUSION: Loss of STRN alters CMs electrical and contractile profiles and affects cell functionality by a disarrangement of Strn-related multi-protein complexes. This leads to impaired microtubules dynamics and Na+ channels trafficking to the plasma membrane, causing a global Na+ and Ca2+ enhancement.


Subject(s)
Calcium , Myocytes, Cardiac , Animals , Myocytes, Cardiac/metabolism , Mice , Calcium/metabolism , Action Potentials/drug effects , Mice, Knockout , Muscle Proteins/metabolism , Muscle Proteins/genetics , Sodium-Calcium Exchanger/metabolism , Sodium-Calcium Exchanger/genetics , Mouse Embryonic Stem Cells/metabolism , Sodium/metabolism
3.
Clin Exp Allergy ; 41(3): 434-42, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20738317

ABSTRACT

BACKGROUND: As an E3 ubiquitin ligase and a molecular adaptor, Cbl-b controls the activation threshold of the antigen receptor and negatively regulates CD28 costimulation, functioning as an intrinsic mediator of T cell anergy that maintains tolerance. However, the role of Cbl-b in the airway immune response to aeroallergens is unclear. OBJECTIVE: To determine the contribution of Cbl-b in tolerance to aeroallergens, we examined ovalbumin (OVA)-induced lung inflammation in Cbl-b-deficient mice. METHODS: Cbl-b(-/-) mice and wild-type (WT) C57BL/6 mice were sensitized and challenged with OVA intranasally, a procedure normally tolerated by WT mice. We analysed lung histology, bronchoalveolar lavage fluid total cell counts and differential, cytokines and chemokines in the airway, and cytokine response by lymphocytes after re-stimulation by OVA antigen. RESULTS: Compared with WT mice, OVA-challenged Cbl-b(-/-) mice showed significantly increased neutrophilic and eosinophilic infiltration in the lung and mucus hyperplasia. The serum levels of IgG2a and IgG1, but not IgE, were increased. The levels of inflammatory mediators IFN-γ, IL-10, IL-12, IL-13, IP-10, MCP-1, MIP-1α, eotaxin, and RANTES, but not IL-17A or IL-6, were elevated in the airway of Cbl-b(-/-) mice. Lymphocytes from Cbl-b(-/-) mice released increased amount of IFN-γ, IL-10, IL-13, and IP-10 in response to OVA re-stimulation. However, no significant changes were noted in the CD4(+) CD25(+) T regulatory cell populations in the lung tissues after OVA stimulation and there was no difference between WT and Cbl-b(-/-) mice. CONCLUSION: These results demonstrate that Cbl-b deficiency leads to a breakdown of tolerance to OVA allergen in the murine airways, probably through increased activation of T effector cells, indicating that Cbl-b is a critical factor in maintaining lung homeostasis upon environmental exposure to aeroallergens.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Allergens/immunology , Immune Tolerance/immunology , Pneumonia/immunology , Proto-Oncogene Proteins c-cbl/immunology , Respiratory Mucosa/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/biosynthesis , Cytokines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Pneumonia/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Respiratory Mucosa/metabolism
4.
Nat Cell Biol ; 3(9): 839-43, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533664

ABSTRACT

Heat-shock protein 70 (Hsp70) has been reported to block apoptosis by binding apoptosis protease activating factor-1 (Apaf-1), thereby preventing constitution of the apoptosome, the Apaf-1/cytochrome c/caspase-9 activation complex [1,2]. Here we show that overexpression of Hsp70 protects Apaf-1-/- cells against death induced by serum withdrawal, indicating that Apaf-1 is not the only target of the anti-apoptotic action of Hsp70. We investigated the effect of Hsp70 on apoptosis mediated by the caspase-independent death effector apoptosis inducing factor (AIF), which is a mitochondrial intermembrane flavoprotein [3,4]. In a cell-free system, Hsp70 prevented the AIF-induced chromatin condensation of purified nuclei. Hsp70 specifically interacted with AIF, as shown by ligand blots and co-immunoprecipitation. Cells overexpressing Hsp70 were protected against the apoptogenic effects of AIF targeted to the extramitochondrial compartment. In contrast, an anti-sense Hsp70 complementary DNA, which reduced the expression of endogenous Hsp70, increased sensitivity to the lethal effect of AIF. The ATP-binding domain of Hsp70 seemed to be dispensable for inhibiting cell death induced by serum withdrawal, AIF binding and AIF inhibition, although it was required for Apaf-1 binding. Together, our data indicate that Hsp70 can inhibit apoptosis by interfering with target proteins other than Apaf-1, one of which is AIF.


Subject(s)
Apoptosis/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Flavoproteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Proteins/metabolism , Animals , Apoptosis Inducing Factor , Apoptotic Protease-Activating Factor 1 , Cell Nucleus/ultrastructure , Cell Survival , Cells, Cultured , Chromatin/physiology , Chromatin/ultrastructure , Culture Media, Serum-Free , Flavoproteins/genetics , Membrane Proteins/genetics , Mice , Proteins/antagonists & inhibitors , Proteins/genetics , Recombinant Proteins/metabolism , Transfection
5.
Nat Med ; 6(4): 429-34, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742150

ABSTRACT

Infections are thought to be important in the pathogenesis of many heart diseases. Coxsackievirus B3 (CVB3) has been linked to chronic dilated cardiomyopathy, a common cause of progressive heart disease, heart failure and sudden death. We show here that the sarcoma (Src) family kinase Lck (p56lck) is required for efficient CVB3 replication in T-cell lines and for viral replication and persistence in vivo. Whereas infection of wild-type mice with human pathogenic CVB3 caused acute and very severe myocarditis, meningitis, hepatitis, pancreatitis and dilated cardiomyopathy, mice lacking the p56lck gene were completely protected from CVB3-induced acute pathogenicity and chronic heart disease. These data identify a previously unknown function of Src family kinases and indicate that p56lck is the essential host factor that controls the replication and pathogenicity of CVB3.


Subject(s)
Cardiomyopathy, Dilated/virology , Coxsackievirus Infections/virology , Enterovirus B, Human/pathogenicity , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Chronic Disease , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/pathology , Encephalomyocarditis virus/pathogenicity , Enterovirus B, Human/physiology , HeLa Cells , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Knockout , Virus Replication , src-Family Kinases/metabolism
6.
J Exp Med ; 172(2): 521-9, 1990 Aug 01.
Article in English | MEDLINE | ID: mdl-2142720

ABSTRACT

Single chicken thymic nurse cells (TNC) placed onto the chorionallantoic membrane (CAM), showed that intra-TNC lymphocytes (TNC-L) possess a strong graft-versus-host reactivity (GVHR) in allogeneic MHC combinations. This reaction shows the morphological, phenotypic, and functional characteristics of a classical GVH reaction (GVHR). The induction of a GVHR was significantly higher for TNC-L as compared with thymocytes or peripheral blood lymphocytes (PBL). The specificity of the GVHR was shown by serial transfer experiments onto appropriate allogeneic and syngeneic secondary embryonic hosts. In immunofluorescence analyses with monoclonal antibodies (mAb) to the chicken alpha/beta and gamma/delta T cell receptors (TCR) and the CD3, CD4, and CD8 equivalents, an enrichment of CD3+/CD4+/CD8- and CD3+/CD-4-/CD8+, TCR-alpha/beta + and TCR- gamma/delta + cells was observed inside TNC as compared with extra-TNC thymocytes. A large proportion of CD4+ and/or CD8+ TCR- gamma/delta + cells were demonstrated inside TNC. A minor population among TCR- gamma/delta extra-TNC thymocytes also expressed CD4 and/or CD8 molecules. Based on functional tests and double staining experiments, we propose that CD4+/CD8+ thymocytes enter the TNC where they may undergo positive selection for MHC restriction and further differentiation to CD4 or CD8 single-positive cells. Taken together these data support the concept that TNC contribute a specialized thymic microenvironment for T cell differentiation and maturation.


Subject(s)
Graft vs Host Reaction/immunology , T-Lymphocytes/immunology , Allantois/immunology , Animals , Antibodies, Monoclonal , Antigens, Differentiation, T-Lymphocyte/analysis , CD3 Complex , CD4 Antigens/analysis , CD8 Antigens , Cells, Cultured , Chick Embryo , Chickens , Chorion/immunology , Fluorescent Antibody Technique , Receptors, Antigen, T-Cell/analysis , T-Lymphocytes/transplantation , Thymus Gland/immunology
7.
J Exp Med ; 189(6): 957-68, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10075979

ABSTRACT

Aberrant activation of cell cycle molecules has been postulated to play a role in apoptosis ("catastrophic cell cycle"). Here we show that in noncycling developing thymocytes, the cyclin- dependent kinase Cdk2 is activated in response to all specific and nonspecific apoptotic stimuli tested, including peptide-specific thymocyte apoptosis. Cdk2 was found to function upstream of the tumor suppressor p53, transactivation of the death promoter Bax, alterations of mitochondrial permeability, Bcl-2, caspase activation, and caspase-dependent proteolytic cleavage of the retinoblastoma protein. Inhibition of Cdk2 completely protected thymocytes from apoptosis, mitochondrial changes, and caspase activation. These data provide the first evidence that Cdk2 activity is crucial for the induction of thymocyte apoptosis.


Subject(s)
Apoptosis , CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/physiology , Protein Serine-Threonine Kinases/physiology , T-Lymphocytes/physiology , Animals , Caspases/physiology , Cyclin-Dependent Kinase 2 , Female , Male , Membrane Potentials , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
8.
J Exp Med ; 176(5): 1459-63, 1992 Nov 01.
Article in English | MEDLINE | ID: mdl-1402689

ABSTRACT

Clonal deletion of thymocytes expressing potentially self-reactive T cell receptors (TCRs) occurs during thymocyte ontogeny. Mice deficient for CD4 expression provide a unique model system to study the contribution of the CD4 molecule in negative selection of T cells reactive against the major histocompatibility complex class II-associated retroviral self-superantigen, Mls-1a. In the presence of Mls-1a determinants, mature CD8+ T cells expressing V beta 6, 8.1, and 9 were deleted in CD4-deficient mice, thus demonstrating that TCR affinity for Mls-1a is sufficient for deletion and that a signal through CD4 was not required. However, in instances where the TCR affinity for Mls-1a is low, as in the case of V beta 7+ T cells, CD4 expression was required for clonal deletion. These results demonstrate that for Mls-1a-mediated clonal deletion of T cells, the requirement for the accessory or coreceptor function of CD4 depends on the affinity of the TCR.


Subject(s)
CD4 Antigens/physiology , Minor Lymphocyte Stimulatory Antigens/immunology , T-Lymphocytes/immunology , Animals , CD4 Antigens/analysis , H-2 Antigens/analysis , Mice , Mice, Inbred CBA , Mice, Inbred DBA , Receptors, Antigen, T-Cell, alpha-beta/analysis
9.
J Exp Med ; 178(5): 1837-42, 1993 Nov 01.
Article in English | MEDLINE | ID: mdl-8228830

ABSTRACT

Experimental induction of most autoimmune diseases appears to depend on the activation of CD4+ T helper cells, while CD8+ lymphocytes may have a role in disease progression. To study the role of CD4+ and CD8+ T cell subsets in T cell-dependent autoimmunity, mice lacking CD4 or CD8 molecules after gene targeting were injected with cardiac myosin to induce organ specific autoimmune myocarditis. Mice homozygous for the CD8 mutation (CD8-/-) developed significantly more severe disease as compared to CD4+/-CD8+/- controls. Surprisingly, CD4-/- mice developed autoimmune myocarditis with infiltration of TCR alpha beta +CD4-CD8- T cells in the heart tissue and appearance of autoantibodies. These data demonstrate that the lack of CD4+ or CD8+ T cells has no significant influence on the initiation of autoimmune myocarditis. CD4+ and CD8+ cells regulate disease severity and these results may explain the occurrence of autoimmunity in CD4 immunodeficiencies.


Subject(s)
Autoimmune Diseases/immunology , CD3 Complex/immunology , CD8 Antigens/immunology , Myocarditis/immunology , Myocardium/immunology , T-Lymphocyte Subsets/immunology , Animals , Autoantibodies/analysis , Autoimmune Diseases/pathology , CD3 Complex/genetics , CD8 Antigens/genetics , Crosses, Genetic , Female , Homozygote , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred Strains , Myocarditis/pathology , Myocardium/pathology , Myosins/immunology
10.
J Exp Med ; 176(6): 1657-63, 1992 Dec 01.
Article in English | MEDLINE | ID: mdl-1460424

ABSTRACT

CD45 is a protein tyrosine phosphatase involved in T and B cell signaling. While peripheral T cells switch CD45 isoforms upon activation, events leading to exon switching during T cell development in the thymus have not been determined. The expression of high molecular weight isoforms of CD45 was examined on thymocytes from nontransgenic and T cell receptor (TCR) transgenic mice. All thymocytes from nontransgenic mice were CD45RB+ as assessed by staining with MB23G2, an anti-CD45RB-specific monoclonal antibody. Interestingly, there was a small population (1-3%) of thymocytes that displayed a higher intensity of staining with MB23G2, CD45RBhigh. CD45RBhigh thymocytes were found in all subsets defined by CD4 and CD8 expression and were also present within the TCR-alpha/beta high population. To analyze whether or not CD45 expression correlated with thymic selection events, expression of CD45RBhigh and a second isoform, CD45RA, was examined on thymocytes from H-Y and 2C TCR transgenic mice and found to correlate with positive and negative selection events but did not occur in nonselecting backgrounds. CD45RA and CD45RBhigh upregulation was also not observed in transgenic mice backcrossed into CD8-deficient mice, a scenario in which there is no positive selection of transgene-expressing thymocytes. These data suggest that modulation of CD45 isoform expression may be involved in thymic selection events.


Subject(s)
Leukocyte Common Antigens/genetics , Protein Tyrosine Phosphatases/genetics , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , Exons , Female , Fluorescent Antibody Technique , Gene Expression , H-2 Antigens/analysis , H-2 Antigens/genetics , Leukocyte Common Antigens/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Mice, Transgenic , Protein Tyrosine Phosphatases/analysis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology
11.
J Exp Med ; 190(9): 1329-42, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10544204

ABSTRACT

The Wiskott-Aldrich syndrome protein (WASp) has been implicated in modulation of lymphocyte activation and cytoskeletal reorganization. To address the mechanisms whereby WASp subserves such functions, we have examined WASp roles in lymphocyte development and activation using mice carrying a WAS null allele (WAS(-)(/)(-)). Enumeration of hemopoietic cells in these animals revealed total numbers of thymocytes, peripheral B and T lymphocytes, and platelets to be significantly diminished relative to wild-type mice. In the thymus, this abnormality was associated with impaired progression from the CD44(-)CD25(+) to the CD44(-)CD25(-) stage of differentiation. WASp-deficient thymocytes and T cells also exhibited impaired proliferation and interleukin (IL)-2 production in response to T cell antigen receptor (TCR) stimulation, but proliferated normally in response to phorbol ester/ionomycin. This defect in TCR signaling was associated with a reduction in TCR-evoked upregulation of the early activation marker CD69 and in TCR-triggered apoptosis. While induction of TCR-zeta, ZAP70, and total protein tyrosine phosphorylation as well as mitogen-activated protein kinase (MAPK) and stress-activated protein/c-Jun NH(2)-terminal kinase (SAPK/JNK) activation appeared normal in TCR-stimulated WAS(-)(/)(-) cells, TCR-evoked increases in intracellular calcium concentration were decreased in WASp-deficient relative to wild-type cells. WAS(-)(/)(-) lymphocytes also manifested a marked reduction in actin polymerization and both antigen receptor capping and endocytosis after TCR stimulation, whereas WAS(-)(/)(-) neutrophils exhibited reduced phagocytic activity. Together, these results provide evidence of roles for WASp in driving lymphocyte development, as well as in the translation of antigen receptor stimulation to proliferative or apoptotic responses, cytokine production, and cytoskeletal rearrangement. The data also reveal a role for WASp in modulating endocytosis and phagocytosis and, accordingly, suggest that the immune deficit conferred by WASp deficiency reflects the disruption of a broad range of cellular behaviors.


Subject(s)
Lymphocyte Activation/immunology , Proteins/genetics , Wiskott-Aldrich Syndrome/immunology , Actins/metabolism , Animals , B-Lymphocytes/immunology , CD3 Complex/immunology , Cell Count , Cell Differentiation , Cytoskeleton/metabolism , Gene Targeting , Immunologic Capping , Interleukin-2/metabolism , Lymph Nodes/immunology , Mice , Mice, Knockout , Neutrophils/immunology , Phagocytosis/immunology , Proteins/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Spleen/immunology , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome Protein
12.
J Exp Med ; 180(5): 1911-20, 1994 Nov 01.
Article in English | MEDLINE | ID: mdl-7964466

ABSTRACT

To reconstitute the human immune system in mice, transgenic mice expressing human CD4 and human major histocompatibility complex (MHC) class II (DQw6) molecules in an endogenous CD4- and CD8-deficient background (mCD4/8-/-), after homologous recombination, have been generated. We report that expression of human CD4 molecule in mCD4/8-/- mice rescues thymocyte development and completely restores the T cell compartment in peripheral lymphoid organs. Upon vesicular stomatitis virus (VSV) challenge, the reconstituted mature T cell population effectively provide T help to B cells in immunoglobulin class switching from IgM to specific IgG-neutralizing antibodies. Human CD4+DQw6+ double transgenic mice are tolerant to DQw6 and the DQw6 molecule functions in antigen presentation, effectively generating a human MHC class II-restricted T cell response to streptococcal M6C2 peptide. These data show that both the hCD4 and DQw6 molecules are functional in mCD4/8-/- mice, fully and stably reconstituting this limb of the human immune system in mice. This animal model provides a powerful in vivo tool to dissect the human CD4-human class II MHC interaction, especially its role in human autoimmune diseases, superantigen-mediated diseases, and acquired immunodeficiency syndrome (AIDS).


Subject(s)
CD4 Antigens/physiology , CD8 Antigens/analysis , HLA-DQ Antigens/physiology , Animals , Antigen Presentation , B-Lymphocytes/physiology , CD4 Antigens/analysis , CD4 Antigens/genetics , HLA-DQ Antigens/genetics , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , T-Lymphocytes/physiology
13.
J Exp Med ; 188(7): 1333-42, 1998 Oct 05.
Article in English | MEDLINE | ID: mdl-9763612

ABSTRACT

Ship is an Src homology 2 domain containing inositol polyphosphate 5-phosphatase which has been implicated as an important signaling molecule in hematopoietic cells. In B cells, Ship becomes associated with Fcgamma receptor IIB (FcgammaRIIB), a low affinity receptor for the Fc portion of immunoglobulin (Ig)G, and is rapidly tyrosine phosphorylated upon B cell antigen receptor (BCR)-FcgammaRIIB coligation. The function of Ship in lymphocytes was investigated in Ship-/- recombination-activating gene (Rag)-/- chimeric mice generated from gene-targeted Ship-/- embryonic stem cells. Ship-/-Rag-/- chimeras showed reduced numbers of B cells and an overall increase in basal serum Ig. Ship-/- splenic B cells displayed prolonged Ca2+ influx, increased proliferation in vitro, and enhanced mitogen-activated protein kinase (MAPK) activation in response to BCR-FcgammaRIIB coligation. These results demonstrate that Ship plays an essential role in FcgammaRIIB-mediated inhibition of BCR signaling, and that Ship is a crucial negative regulator of Ca2+ flux and MAPK activation.


Subject(s)
B-Lymphocytes/metabolism , Phosphoric Monoester Hydrolases/physiology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Animals , B-Lymphocytes/immunology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cytokines/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Immunoglobulins/blood , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1 , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Receptors, IgG/metabolism , T-Lymphocytes/cytology , Th1 Cells/metabolism , Th2 Cells/metabolism , Vesicular stomatitis Indiana virus/immunology
14.
J Exp Med ; 192(8): 1081-92, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-11034598

ABSTRACT

Syncytia arising from the fusion of cells expressing a lymphotropic HIV type 1-encoded envelope glycoprotein complex (Env) with cells expressing the CD4/CXC chemokine receptor 4 complex spontaneously undergo cell death. Here we show that this process is accompanied by caspase activation and signs of mitochondrial membrane permeabilization (MMP), including the release of intermembrane proteins such as cytochrome c (Cyt-c) and apoptosis-inducing factor (AIF) from mitochondria. In Env-induced syncytia, caspase inhibition did not suppress AIF- and Cyt-c translocation, yet it prevented all signs of nuclear apoptosis. Translocation of Bax to mitochondria led to MMP, which was inhibited by microinjected Bcl-2 protein or bcl-2 transfection. Bcl-2 also prevented the subsequent nuclear chromatin condensation and DNA fragmentation. The release of AIF occurred before that of Cyt-c and before caspase activation. Microinjection of AIF into syncytia sufficed to trigger rapid, caspase-independent Cyt-c release. Neutralization of endogenous AIF by injection of an antibody prevented all signs of spontaneous apoptosis occurring in syncytia, including the Cyt-c release and nuclear apoptosis. In contrast, Cyt-c neutralization only prevented nuclear apoptosis, and did not affect AIF release. Our results establish that the following molecular sequence governs apoptosis of Env-induced syncytia: Bax-mediated/Bcl-2-inhibited MMP --> AIF release --> Cyt-c release --> caspase activation --> nuclear apoptosis.


Subject(s)
Apoptosis/physiology , CD4 Antigens/physiology , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Gene Products, env/metabolism , Giant Cells/virology , HIV-1/physiology , Mitochondria/physiology , CD4-Positive T-Lymphocytes/physiology , Cell Fusion , Coculture Techniques , Genes, env , Giant Cells/cytology , Giant Cells/physiology , HeLa Cells , Humans , Intracellular Membranes/physiology , Kinetics , Permeability , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection
15.
J Exp Med ; 188(11): 2099-111, 1998 Dec 07.
Article in English | MEDLINE | ID: mdl-9841924

ABSTRACT

The protooncogene Vav functions as a GDP/GTP exchange factor (GEF) for Rho-like small GTPases involved in cytoskeletal reorganization and cytokine production in T cells. Gene-targeted mice lacking Vav have a severe defect in positive and negative selection of T cell antigen receptor transgenic thymocytes in vivo, and vav-/- thymocytes are completely resistant to peptide-specific and anti-CD3/anti-CD28-mediated apoptosis. Vav acts upstream of mitochondrial pore opening and caspase activation. Biochemically, Vav regulates peptide-specific Ca2+ mobilization and actin polymerization. Peptide-specific cell death was blocked both by cytochalasin D inhibition of actin polymerization and by inhibition of protein kinase C (PKC). Activation of PKC with phorbol ester restored peptide-specific apoptosis in vav-/- thymocytes. Vav was found to bind constitutively to PKC-theta in thymocytes. Our results indicate that peptide-triggered thymocyte apoptosis is mediated via Vav activation, changes in the actin cytoskeleton, and subsequent activation of a PKC isoform.


Subject(s)
Apoptosis/genetics , Apoptosis/immunology , Cell Cycle Proteins , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , CD28 Antigens/immunology , CD3 Complex/immunology , Cytokines/immunology , Cytoskeleton/immunology , Cytoskeleton/pathology , Mice , Mice, Transgenic , Peptides , Proto-Oncogene Proteins c-vav , Signal Transduction/immunology
16.
J Exp Med ; 194(6): 757-68, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11560992

ABSTRACT

The dual specificity kinases mitogen-activated protein kinase (MAPK) kinase (MKK)7 and MKK4 are the only molecules known to directly activate the stress kinases stress-activated protein kinases (SAPKs)/c-Jun N-terminal kinases (JNKs) in response to environmental or mitogenic stimuli. To examine the physiological role of MKK7 in hematopoietic cells, we used a gene targeting strategy to mutate MKK7 in murine T and B cells and non-lymphoid mast cells. Loss of MKK7 in thymocytes and mature B cells results in hyperproliferation in response to growth factor and antigen receptor stimulation and increased thymic cellularity. Mutation of mkk7 in mast cells resulted in hyperproliferation in response to the cytokines interleukin (IL)-3 and stem cell factor (SCF). SAPK/JNK activation was completely abolished in the absence of MKK7, even though expression of MKK4 was strongly upregulated in mkk7(-/-) mast cell lines, and phosphorylation of MKK4 occurred normally in response to multiple stress stimuli. Loss of MKK7 did not affect activation of extracellular signal-regulated kinase (ERK)1/2 or p38 MAPK. mkk7(-/-) mast cells display reduced expression of JunB and the cell cycle inhibitor p16INK4a and upregulation of cyclinD1. Reexpression of p16INK4a in mkk7(-/-) mast cells abrogates the hyperproliferative response. Apoptotic responses to a variety of stimuli were not affected. Thus, MKK7 is an essential and specific regulator of stress-induced SAPK/JNK activation in mast cells and MKK7 negatively regulates growth factor and antigen receptor-driven proliferation in hematopoietic cells. These results indicate that the MKK7-regulated stress signaling pathway can function as negative regulator of cell growth in multiple hematopoietic lineages.


Subject(s)
B-Lymphocytes/cytology , MAP Kinase Kinase 4 , Mast Cells/cytology , Mitogen-Activated Protein Kinase Kinases/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Growth Factor/metabolism , T-Lymphocytes/cytology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Division , Enzyme Activation , Gene Targeting , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Interleukin-3/metabolism , Interleukin-3/pharmacology , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 7 , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Thymus Gland/cytology
17.
J Exp Med ; 192(4): 571-80, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10952727

ABSTRACT

Apaf-1(-/-) or caspase-3(-/-) cells treated with a variety of apoptosis inducers manifest apoptosis-associated alterations including the translocation of apoptosis-inducing factor (AIF) from mitochondria to nuclei, large scale DNA fragmentation, and initial chromatin condensation (stage I). However, when compared with normal control cells, Apaf-1(-/-) or caspase-3(-/-) cells fail to exhibit oligonucleosomal chromatin digestion and a more advanced pattern of chromatin condensation (stage II). Microinjection of such cells with recombinant AIF only causes peripheral chromatin condensation (stage I), whereas microinjection with activated caspase-3 or its downstream target caspase-activated DNAse (CAD) causes a more pronounced type of chromatin condensation (stage II). Similarly, when added to purified HeLa nuclei, AIF causes stage I chromatin condensation and large-scale DNA fragmentation, whereas CAD induces stage II chromatin condensation and oligonucleosomal DNA degradation. Furthermore, in a cell-free system, concomitant neutralization of AIF and CAD is required to suppress the nuclear DNA loss caused by cytoplasmic extracts from apoptotic wild-type cells. In contrast, AIF depletion alone suffices to suppress the nuclear DNA loss contained in extracts from apoptotic Apaf-1(-/-) or caspase-3(-/-) cells. As a result, at least two redundant parallel pathways may lead to chromatin processing during apoptosis. One of these pathways involves Apaf-1 and caspases, as well as CAD, and leads to oligonucleosomal DNA fragmentation and advanced chromatin condensation. The other pathway, which is caspase-independent, involves AIF and leads to large-scale DNA fragmentation and peripheral chromatin condensation.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cell Nucleus/metabolism , Flavoproteins/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis Inducing Factor , Apoptotic Protease-Activating Factor 1 , Arsenites/pharmacology , Caspase 3 , Caspases/genetics , Cells, Cultured , Cisplatin/pharmacology , Cytochrome c Group/genetics , Cytochrome c Group/metabolism , DNA Fragmentation , Deoxyribonucleases/genetics , Deoxyribonucleases/metabolism , Etoposide/pharmacology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Flavoproteins/genetics , Fluorescent Dyes/metabolism , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Microinjections , Proteins/genetics , Recombinant Proteins/metabolism
18.
J Exp Med ; 186(6): 941-53, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9294148

ABSTRACT

The dual specific kinase SAPK/ERK1 kinase (SEK1; mitogen-activated protein kinase kinase 4/Jun NH2 terminal kinase [ JNK] kinase) is a direct activator of stress-activated protein kinases ([SAPKs]/JNKs) in response to CD28 costimulation, CD40 signaling, or activation of the germinal center kinase. Here we show that SEK1(-/-) recombination-activating gene (RAG)2(-/-) chimeric mice have a partial block in B cell maturation. However, peripheral B cells displayed normal responses to IL-4, IgM, and CD40 cross-linking. SEK1(-/-) peripheral T cells showed decreased proliferation and IL-2 production after CD28 costimulation and PMA/Ca2+ ionophore activation. Although CD28 expression was absolutely crucial to generate vesicular stomatitis virus (VSV)-specific germinal centers, SEK1(-/-)RAG2(-/-) chimeras mounted a protective antiviral B cell response, exhibited normal IgG class switching, and made germinal centers in response to VSV. Interestingly, PMA/Ca2+ ionophore stimulation, which mimics TCR-CD3 and CD28-mediated signal transduction, induced SAPK/JNK activation in peripheral T cells, but not in thymocytes, from SEK1(-/-) mice. These results show that signaling pathways for SAPK activation are developmentally regulated in T cells. Although SEK1(-/-) thymocytes failed to induce SAPK/JNK in response to PMA/Ca2+ ionophore, SEK1(-/-)RAG2(-/-) thymocytes proliferated and made IL-2 after PMA/Ca2+ ionophore and CD3/CD28 stimulation, albeit at significantly lower levels compared to SEK1(+/+)RAG2(-/-) thymocytes, implying that CD28 costimulation and PMA/Ca2+ ionophore-triggered signaling pathways exist that can mediate proliferation and IL-2 production independently of SAPK activation. Our data provide the first genetic evidence that SEK1 is an important effector molecule that relays CD28 signaling to IL-2 production and T cell proliferation.


Subject(s)
CD28 Antigens/metabolism , Interleukin-2/biosynthesis , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Kinases/deficiency , Protein Serine-Threonine Kinases/deficiency , Protein-Tyrosine Kinases/deficiency , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation , Chimera , DNA Primers/genetics , Germinal Center/cytology , Germinal Center/immunology , Immunoglobulin Switch Region , JNK Mitogen-Activated Protein Kinases , Lymphocyte Activation , Mice , Mice, Knockout , Polymerase Chain Reaction , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Recombination, Genetic , Signal Transduction , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/pathogenicity
19.
Cell Death Differ ; 15(1): 89-93, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17762881

ABSTRACT

During the development and organogenesis of all multicellular organisms, cell fate decisions determine whether cells undergo proliferation, differentiation, or aging. Two independent stress kinase signaling pathways, p38-MAPK, and JNKs, have evolved that relay developmental and environmental cues to determine cell responses. Although multiple stimuli can activate these two stress kinase pathways, the functional interactions and molecular cross-talks between these common second signaling cascades are poorly elucidated. Here we report that JNK and p38-MAPK pathways antagonistically control cellular senescence, oncogenic transformation, and proliferation in primary mouse embryonic fibroblasts (MEFs). Similarly, genetic inactivation of the JNK pathway results in impaired proliferation of fetal hepatoblasts in vitro and defective adult liver regeneration in vivo, which is rescued by inhibition of the p38-MAPK pathway. Thus, the balance between the two stress-signaling pathways, MKK7-JNK and MKK3/6-p38-MAPK, determines cell fate and links environmental and developmental stress to cell cycle arrest, senescence, oncogenic transformation, and adult tissue regeneration.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic , Cellular Senescence , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Regeneration , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cells, Cultured , Fibroblasts/metabolism , Hepatocytes/metabolism , Mice , Mice, Mutant Strains
20.
Cell Death Differ ; 15(7): 1113-23, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18309324

ABSTRACT

As a result of the genetic experiments performed in Caenorhabditis elegans, it has been tacitly assumed that the core proteins of the 'apoptotic machinery' (CED-3, -4, -9 and EGL-1) would be solely involved in cell death regulation/execution and would not exert any functions outside of the cell death realm. However, multiple studies indicate that the mammalian orthologs of these C. elegans proteins (i.e. caspases, Apaf-1 and multidomain proteins of the Bcl-2 family) participate in cell death-unrelated processes. Similarly, loss-of-function mutations of ced-4 compromise the mitotic arrest of DNA-damaged germline cells from adult nematodes, even in a context in which the apoptotic machinery is inoperative (for instance due to mutations of egl-1 or ced-3). Moreover, EGL-1 is required for the activation of autophagy in starved nematodes. Finally, the depletion of caspase-independent death effectors, such as apoptosis-inducing factor (AIF) and endonuclease G, provokes cell death-independent consequences, both in mammals and in yeast (Saccharomyces cerevisiae). These results corroborate the conjecture that any kind of protein that has previously been specifically implicated in apoptosis might have a phylogenetically conserved apoptosis-unrelated function, most likely as part of an adaptive response to cellular stress.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Signal Transduction , Adaptation, Physiological , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Caspases/metabolism , Evolution, Molecular , Humans , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proteins/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL