Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 22(11): 1837-45, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25882048

ABSTRACT

Isocitrate dehydrogenase-1 (Idh1) is an important metabolic enzyme that produces NADPH by converting isocitrate to α-ketoglutarate. Idh1 is known to reduce reactive oxygen species (ROS) induced in cells by treatment with lipopolysaccharide (LPS) in vitro. Here, we used Idh1-deficient knockout (Idh1 KO) mice to investigate the role of Idh1 in antioxidant defense in vivo. Idh1 KO mice showed heightened susceptibility to death induced by LPS and exhibited increased serum levels of inflammatory cytokines such as tumor necrosis factor-α and interleukin-6. The serum of LPS-injected Idh1 KO mice also contained elevated levels of AST, a marker of inflammatory liver damage. Furthermore, after LPS injection, livers of Idh1 KO mice showed histological evidence of elevated oxidative DNA damage compared with livers of wild-type (WT) mice. Idh1 KO livers showed a faster and more pronounced oxidative stress than WT livers. In line with that, Idh1 KO hepatocytes showed higher ROS levels and an increase in the NADP(+)/NADPH ratio when compared with hepatocytes isolated from WT mice. These results suggest that Idh1 has a physiological function in protecting cells from oxidative stress by regulating the intracellular NADP(+)/NADPH ratio. Our findings suggest that stimulation of Idh1 activity may be an effective therapeutic strategy for reducing oxidative stress during inflammatory responses, including the early stages of septic shock.


Subject(s)
Endotoxins/pharmacology , Hepatocytes/drug effects , Hepatocytes/enzymology , Isocitrate Dehydrogenase/metabolism , NADP/metabolism , Animals , Cells, Cultured , Flow Cytometry , Isocitrate Dehydrogenase/genetics , Mice , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
Cell Death Differ ; 22(1): 164-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25257173

ABSTRACT

During virus infection and autoimmune disease, inflammatory dendritic cells (iDCs) differentiate from blood monocytes and infiltrate infected tissue. Following acute infection with hepatotropic viruses, iDCs are essential for re-stimulating virus-specific CD8(+) T cells and therefore contribute to virus control. Here we used the lymphocytic choriomeningitis virus (LCMV) model system to identify novel signals, which influence the recruitment and activation of iDCs in the liver. We observed that intrinsic expression of Toso (Faim3, FcµR) influenced the differentiation and activation of iDCs in vivo and DCs in vitro. Lack of iDCs in Toso-deficient (Toso(-/-)) mice reduced CD8(+) T-cell function in the liver and resulted in virus persistence. Furthermore, Toso(-/-) DCs failed to induce autoimmune diabetes in the rat insulin promoter-glycoprotein (RIP-GP) autoimmune diabetes model. In conclusion, we found that Toso has an essential role in the differentiation and maturation of iDCs, a process that is required for the control of persistence-prone virus infection.


Subject(s)
Carrier Proteins/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/genetics , Cell Differentiation/genetics , Dendritic Cells/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Immunity, Cellular , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Liver/immunology , Liver/pathology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Rats
3.
Blood ; 97(6): 1854-60, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238129

ABSTRACT

Studies with neutralizing antibodies have indicated roles for platelet-endothelial cell adhesion molecule-1 (PECAM-1) in leukocyte migration through the endothelium and the perivascular basement membrane. Because some of these findings have been contentious, this study aimed to explore the role of PECAM-1 in leukocyte migration by analyzing leukocyte responses in interleukin 1beta (IL-1beta)- and tumor necrosis factor-alpha (TNFalpha)-activated cremasteric venules of PECAM-1-deficient mice using intravital and electron microscopy. Although no differences in levels of leukocyte rolling flux or firm adhesion were observed, a delay in leukocyte transmigration in response to IL-1beta, but not TNFalpha, was detected in PECAM-1-deficient mice. Electron microscopy indicated that this delay occurred at the level of perivascular basement membrane. To address the cytokine specificity of PECAM-1 dependence, in vitro experiments demonstrated that TNFalpha, but not IL-1beta, could induce rapid adhesion of murine neutrophils to protein-coated surfaces, suggesting that TNFalpha elicited leukocyte transmigration in wild-type mice via direct stimulation of leukocytes. In summary, the results suggest a regulatory role for PECAM-1 in leukocyte migration through the perivascular basement membrane, a role that appears to be cytokine-specific and associated with the ability of the cytokine to stimulate rapid neutrophil adhesion.


Subject(s)
Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Basement Membrane , Cell Adhesion/drug effects , Chemotaxis, Leukocyte/drug effects , Interleukin-1/pharmacology , Leukocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Neutrophil Activation/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Venules/physiology
4.
Cell ; 104(1): 33-42, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11163238

ABSTRACT

Bcl10, a CARD-containing protein identified from the t(1;14)(p22;q32) breakpoint in MALT lymphomas, has been shown to induce apoptosis and activate NF-kappaB in vitro. We show that one-third of bcl10-/- embryos developed exencephaly, leading to embryonic lethality. Surprisingly, bcl10-/- cells retained susceptibility to various apoptotic stimuli in vivo and in vitro. However, surviving bcl10-/- mice were severely immunodeficient and bcl10-/- lymphocytes are defective in antigen receptor or PMA/Ionomycin-induced activation. Early tyrosine phosphorylation, MAPK and AP-1 activation, and Ca2+ signaling were normal in mutant lymphocytes, but antigen receptor-induced NF-kappaB activation was absent. Thus, Bcl10 functions as a positive regulator of lymphocyte proliferation that specifically connects antigen receptor signaling in B and T cells to NF-kappaB activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Central Nervous System/abnormalities , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neural Tube Defects/physiopathology , Receptors, Antigen/metabolism , Animals , Anisomycin/pharmacology , Antibody Formation/immunology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Apoptosis/radiation effects , B-Cell CLL-Lymphoma 10 Protein , B-Lymphocytes/immunology , Cell Division/immunology , Central Nervous System/physiology , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Genes, Lethal/immunology , Immunity, Cellular/immunology , Lymphocyte Activation/physiology , Mice , Mice, Knockout , NF-kappa B/immunology , Neoplasm Proteins/metabolism , Neural Tube Defects/immunology , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Receptors, Antigen/immunology , Signal Transduction/immunology , Staurosporine/pharmacology , T-Lymphocytes/immunology
5.
J Exp Med ; 192(3): 325-36, 2000 Aug 07.
Article in English | MEDLINE | ID: mdl-10934221

ABSTRACT

Interferon (IFN) regulatory factor (IRF)-2 was originally described as an antagonist of IRF-1-mediated transcriptional regulation of IFN-inducible genes. IRF-1(-/)- mice exhibit defective T helper type 1 (Th1) cell differentiation. We have used experimental leishmaniasis to show that, like IRF-1(-/)- mice, IRF-2(-/)- mice are susceptible to Leishmania major infection due to a defect in Th1 differentiation. Natural killer (NK) cell development is compromised in both IRF-1(-/)- and IRF-2(-/)- mice, but the underlying mechanism differs. NK (but not NK(+) T) cell numbers are decreased in IRF-2(-/)- mice, and the NK cells that are present are immature in phenotype. Therefore, like IRF-1, IRF-2 is required for normal generation of Th1 responses and for NK cell development in vivo. In this particular circumstance the absence of IRF-2 cannot be compensated for by the presence of IRF-1 alone. Mechanistically, IRF-2 may act as a functional agonist rather than antagonist of IRF-1 for some, but not all, IFN-stimulated regulatory element (ISRE)-responsive genes.


Subject(s)
DNA-Binding Proteins/physiology , Killer Cells, Natural/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Repressor Proteins , Th1 Cells/immunology , Transcription Factors/physiology , Animals , Bone Marrow/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , DNA-Binding Proteins/genetics , Disease Models, Animal , Disease Susceptibility/immunology , Female , Interferon Regulatory Factor-2 , Interleukin-12/biosynthesis , Interleukin-15/immunology , Killer Cells, Natural/cytology , Leishmaniasis, Cutaneous/blood , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Th1 Cells/cytology
6.
Nat Immunol ; 1(1): 77-82, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10881179

ABSTRACT

Brca1 (breast cancerl, early onset) deficiency results in early embryonic lethality. As Brca1 is highly expressed in the T cell lineage, a T cell-specific disruption of Brca1 was generated to assess the role of Brca1 in relation to T lymphocyte development. We found that thymocyte development in Brca1-/- mice was impaired not as a result of V(D)J T cell receptor (TCR) recombination but because thymocytes had increased expression of tumor protein p53. Chromosomal damage accumulation and abnormal cell death were observed in mutant cells. We found that cell death inhibitor Bcl-2 overexpression, or p53-/- backgrounds, completely restored survival and development of Brca1-/- thymocytes; peripheral T cell numbers were not totally restored in Brcal-/- p53-/- mice; and that a mutant background for p21 (cyclin-dependent kinase inhibitor 1A) did not restore Brca1-/- thymocyte development, but partially restored peripheral T cell development. Thus, the outcome of Brca1 deficiency was dependent on cellular context, with the major defects being increased apoptosis in thymocytes, and defective proliferation in peripheral T cells.


Subject(s)
BRCA1 Protein/genetics , BRCA1 Protein/immunology , Gene Rearrangement, T-Lymphocyte/immunology , T-Lymphocytes/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation/immunology , Mice , Mice, Knockout , T-Lymphocytes/cytology
7.
J Immunol ; 163(9): 4909-16, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10528193

ABSTRACT

Caspase-3 is essential for Fas-mediated apoptosis in vitro. We investigated the role of caspase-3 in Fas-mediated cell death in vivo by injecting caspase-3-deficient mice with agonistic anti-Fas Ab. Wild-type controls died rapidly of fulminant hepatitis, whereas the survival of caspase-3-/- mice was increased due to a delay in hepatocyte cell death. Bcl-2 expression in the liver was dramatically decreased in wild-type mice following anti-Fas injection, but was unchanged in caspase-3-/- mice. Hepatocytes from anti-Fas-injected wild-type, but not caspase-3-/-, mice released cytochrome c into the cytoplasm. Western blotting confirmed the lack of caspase-3-mediated cleavage of Bcl-2. Presumably the presence of intact Bcl-2 in caspase-3-/- hepatocytes prevents the release of cytochrome c from the mitochondria, a required step for the mitochondrial death pathway. We also show by Western blot that Bcl-xL, caspase-9, caspase-8, and Bid are processed by caspase-3 in injected wild-type mice but that this processing does not occur in caspase-3-/- mice. This study thus provides novel in vivo evidence that caspase-3, conventionally known for its downstream effector function in apoptosis, also modifies Bcl-2 and other upstream proteins involved in the regulation of Fas-mediated apoptosis.


Subject(s)
Apoptosis/immunology , Caspases/physiology , Liver/enzymology , Liver/immunology , fas Receptor/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Caspase 3 , Caspases/genetics , Cytochrome c Group/metabolism , In Situ Nick-End Labeling , Injections, Intraperitoneal , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational/immunology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Staining and Labeling , Survival Analysis , fas Receptor/immunology
8.
Immunity ; 11(3): 379-89, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10514016

ABSTRACT

Tumor necrosis factor (TNF) receptor-associated factor 2 (TRAF2) can interact with various members of the TNF receptor family. Previously, we reported that TRAF2-deficient mice die prematurely and have elevated serum TNF levels. In this study, we demonstrate that TRAF2-deficient macrophages produce increased amounts of nitric oxide (NO) and TNF in response to TNF stimulation. Furthermore, we could enhance the survival of TRAF2-deficient mice by eliminating either TNF or TNFR1. Using these double-knockout mice, we show that in the absence of TRAF2, the T helper-dependent antibody response, CD40-mediated proliferation, and NF-kappaB activation are defective. These data demonstrate two important roles of TRAF2, one as a negative regulator of certain TNFR1 signals and the other as a positive mediator of CD40 signaling.


Subject(s)
Antigens, CD/metabolism , CD40 Antigens/metabolism , Proteins/physiology , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Animals , Cell Division , Cells, Cultured , Female , Immunoglobulin Class Switching , Immunoglobulin Isotypes , Interleukin-12/biosynthesis , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phenotype , Proteins/genetics , Receptors, Tumor Necrosis Factor, Type I , Spleen/cytology , TNF Receptor-Associated Factor 2 , Tumor Necrosis Factor-alpha/biosynthesis , Vesicular stomatitis Indiana virus
9.
Genes Dev ; 13(8): 1015-24, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10215628

ABSTRACT

Bone resorption and remodeling is an intricately controlled, physiological process that requires the function of osteoclasts. The processes governing both the differentiation and activation of osteoclasts involve signals induced by osteoprotegerin ligand (OPGL), a member of tumor necrosis factor (TNF) superfamily, and its cognate receptor RANK. The molecular mechanisms of the intracellular signal transduction remain to be elucidated. Here we report that mice deficient in TNF receptor-associated factor 6 (TRAF6) are osteopetrotic with defects in bone remodeling and tooth eruption due to impaired osteoclast function. Using in vitro assays, we demonstrate that TRAF6 is crucial not only in IL-1 and CD40 signaling but also, surprisingly, in LPS signaling. Furthermore, like TRAF2 and TRAF3, TRAF6 is essential for perinatal and postnatal survival. These findings establish unexpectedly diverse and critical roles for TRAF6 in perinatal and postnatal survival, bone metabolism, LPS, and cytokine signaling.


Subject(s)
CD40 Antigens/metabolism , Interleukin-1/metabolism , Lipopolysaccharides/metabolism , Mitogen-Activated Protein Kinases , Osteopetrosis/physiopathology , Proteins/physiology , Signal Transduction , Animals , B-Lymphocytes/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Enzyme Activation , Female , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Mice , Mice, Knockout , NF-kappa B/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Proteins/genetics , TNF Receptor-Associated Factor 6
10.
J Immunol ; 162(5): 3022-30, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10072554

ABSTRACT

Platelet/endothelial cell adhesion molecule-1 (PECAM-1; CD31), a member of the Ig superfamily, is expressed strongly at endothelial cell-cell junctions, on platelets, and on most leukocytes. CD31 has been postulated to play a role in vasculogenesis and angiogenesis, and has been implicated as a key mediator of the transendothelial migration of leukocytes. To further define the physiologic role of CD31, we used targeted gene disruption of the CD31 gene in embryonic stem cells to generate CD31-deficient mice. CD31-deficient mice (CD31KO) are viable and born at the expected Mendelian frequency, remain healthy, and exhibit no obvious vascular developmental defects. In response to inflammatory challenge, polymorphonuclear leukocytes of CD31KO mice are arrested between the vascular endothelium and the basement membrane of inflammatory site mesenteric microvessels, confirming a role for CD31 in the migration of neutrophils through the subendothelial extracellular matrix. Normal numbers of leukocytes are recovered from inflammatory sites in CD31KO mice, however, suggesting that the defect in leukocyte migration across basal lamina observed in the absence of CD31 may be compensated for by the use of other adhesion molecules, or possibly an increased rate of migration. Homing of T lymphocytes in vivo is normal, and CD31KO mice are able to mount a cutaneous hypersensitivity response normally. In addition, CD31-mediated homophilic adhesion does not appear to play a role in platelet aggregation in vitro. This study provides genetic evidence that CD31 is involved in transbasement membrane migration, but does not play an obligatory role in either vascular development or leukocyte migration.


Subject(s)
Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Blood Cell Count , Blood Platelets/physiology , Cell Movement , Female , Leukocytes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/physiology
11.
Cell ; 94(3): 339-52, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9708736

ABSTRACT

Mutation of Caspase 9 (Casp9) results in embryonic lethality and defective brain development associated with decreased apoptosis. Casp9-/- embryonic stem cells and embryonic fibroblasts are resistant to several apoptotic stimuli, including UV and gamma irradiation. Casp9-/- thymocytes are also resistant to dexamethasone- and gamma irradiation-induced apoptosis, but are surprisingly sensitive to apoptosis induced by UV irradiation or anti-CD95. Resistance to apoptosis is accompanied by retention of the mitochondrial membrane potential in mutant cells. In addition, cytochrome c is translocated to the cytosol of Casp9-/- ES cells upon UV stimulation, suggesting that Casp9 acts downstream of cytochrome c. Caspase processing is inhibited in Casp9-/- ES cells but not in thymocytes or splenocytes. Comparison of the requirement for Casp9 and Casp3 in different apoptotic settings indicates the existence of at least four different apoptotic pathways in mammalian cells.


Subject(s)
Apoptosis/genetics , Caspases , Cysteine Endopeptidases/physiology , Signal Transduction/genetics , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 9 , Cell Line , Cerebral Cortex/abnormalities , Cysteine Endopeptidases/genetics , Cytochrome c Group/metabolism , Dexamethasone/pharmacology , Embryo, Mammalian , Enzyme Activation/genetics , Fibroblasts/cytology , Fibroblasts/enzymology , Gamma Rays , Gene Expression Regulation, Developmental , Lymphocyte Activation , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mitochondria/enzymology , Organ Specificity/genetics , Prosencephalon/abnormalities , Spleen/cytology , Spleen/immunology , Spleen/radiation effects , Stem Cells , Thymus Gland/cytology , Thymus Gland/enzymology
12.
J Exp Med ; 187(6): 967-72, 1998 Mar 16.
Article in English | MEDLINE | ID: mdl-9500799

ABSTRACT

In contrast to conventional T cells, natural killer (NK) 1.1+ T cell receptor (TCR)-alpha/beta+ (NK1+T) cells, NK cells, and intestinal intraepithelial lymphocytes (IELs) bearing CD8-alpha/alpha chains constitutively express the interleukin (IL)-2 receptor (R)beta/15Rbeta chain. Recent studies have indicated that IL-2Rbeta/15Rbeta chain is required for the development of these lymphocyte subsets, outlining the importance of IL-15. In this study, we investigated the development of these lymphocyte subsets in interferon regulatory factor 1-deficient (IRF-1-/-) mice. Surprisingly, all of these lymphocyte subsets were severely reduced in IRF-1-/- mice. Within CD8-alpha/alpha+ intestinal IEL subset, TCR-gamma/delta+ cells and TCR-alpha/beta+ cells were equally affected by IRF gene disruption. In contrast to intestinal TCR-gamma/delta+ cells, thymic TCR-gamma/delta+ cells developed normally in IRF-1-/- mice. Northern blot analysis further revealed that the induction of IL-15 messenger RNA was impaired in IRF-1-/- bone marrow cells, and the recovery of these lymphocyte subsets was observed when IRF-1-/- cells were cultured with IL-15 in vitro. These data indicate that IRF-1 regulates IL-15 gene expression, which may control the development of NK1+T cells, NK cells, and CD8-alpha/alpha+ IELs.


Subject(s)
Antigens/analysis , DNA-Binding Proteins/physiology , Intestinal Mucosa/immunology , Killer Cells, Natural/physiology , Phosphoproteins/physiology , Proteins/analysis , Receptors, Antigen, T-Cell, alpha-beta/analysis , T-Lymphocyte Subsets/physiology , Transcription Factors/physiology , Animals , Antigens, Ly , Antigens, Surface , Gene Expression Regulation , Interferon Regulatory Factor-1 , Interleukin-15/genetics , Lectins, C-Type , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B , RNA, Messenger/analysis , Receptors, Antigen, T-Cell, gamma-delta/analysis
13.
Genes Dev ; 12(6): 806-19, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9512515

ABSTRACT

Caspases are fundamental components of the mammalian apoptotic machinery, but the precise contribution of individual caspases is controversial. CPP32 (caspase 3) is a prototypical caspase that becomes activated during apoptosis. In this study, we took a comprehensive approach to examining the role of CPP32 in apoptosis using mice, embryonic stem (ES) cells, and mouse embryonic fibroblasts (MEFs) deficient for CPP32. CPP32(ex3-/-) mice have reduced viability and, consistent with an earlier report, display defective neuronal apoptosis and neurological defects. Inactivation of CPP32 dramatically reduces apoptosis in diverse settings, including activation-induced cell death (AICD) of peripheral T cells, as well as chemotherapy-induced apoptosis of oncogenically transformed CPP32(-/-) MEFs. As well, the requirement for CPP32 can be remarkably stimulus-dependent: In ES cells, CPP32 is necessary for efficient apoptosis following UV- but not gamma-irradiation. Conversely, the same stimulus can show a tissue-specific dependence on CPP32: Hence, TNFalpha treatment induces normal levels of apoptosis in CPP32 deficient thymocytes, but defective apoptosis in oncogenically transformed MEFs. Finally, in some settings, CPP32 is required for certain apoptotic events but not others: Select CPP32(ex3-/-) cell types undergoing cell death are incapable of chromatin condensation and DNA degradation, but display other hallmarks of apoptosis. Together, these results indicate that CPP32 is an essential component in apoptotic events that is remarkably system- and stimulus-dependent. Consequently, drugs that inhibit CPP32 may preferentially disrupt specific forms of cell death.


Subject(s)
Apoptosis/physiology , Caspases , Cell Nucleus/metabolism , Cysteine Endopeptidases/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , CD3 Complex/pharmacology , Caspase 3 , Cell Death/physiology , Cell Division/physiology , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/physiology , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Embryonic and Fetal Development , Female , Gene Expression/genetics , Gene Expression/physiology , Longevity/genetics , Longevity/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Mutation/genetics , Mutation/physiology , Neutrophils/physiology , Osmotic Pressure , Stem Cells/radiation effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology , Tumor Cells, Cultured/radiation effects , Ultraviolet Rays , fas Receptor/pharmacology
14.
Circulation ; 96(2): 585-91, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9244230

ABSTRACT

BACKGROUND: Production of NO by inducible NO synthase (iNOS) has been implicated in the pathology of spontaneous and antigen-induced autoimmune diseases, and iNOS is expressed in the myocardium of patients with heart failure. It is not clear whether inflammatory murine autoimmune heart disease, an experimental model for human postviral heart disease, is characterized by increased iNOS expression within the heart and whether iNOS and NO are essential in the pathogenesis of autoimmune myocarditis. METHODS AND RESULTS: In the murine model of cardiac myosin-induced myocarditis, we demonstrate that iNOS expression was elicited in inflammatory macrophages and in distinct cardiomyocytes. Autoimmune heart disease was accompanied by formation of the NO reaction product nitrotyrosine in inflammatory macrophages as well as in cardiomyocytes. iNOS expression and nitrotyrosine formation were strictly dependent on myocardial inflammation. Focal myocarditis was sufficient to induce nitrotyrosine formation throughout the whole heart muscle. Mice defective for the interferon regulatory transcription factor-1 (IRF-1(-/-)) after gene targeting failed to induce iNOS expression and nitrotyrosine formation in the heart but developed cardiac myosin-induced myocarditis at prevalence and severity similar to those of heterozygous littermates (IRF-1(+/-)). CONCLUSIONS: These data provide the first in vivo evidence that iNOS expression and NO synthesis in macrophages and distinct cardiomyocytes are elicited in experimental murine inflammatory heart disease. The transcription factor IRF-1 controls iNOS expression and NO synthesis in disease. Because autoimmune myocarditis can develop in animals lacking IRF-1, these mice will be useful to elucidate the link between iNOS expression in inflammatory heart disease and the development of dilated cardiomyopathy and heart failure.


Subject(s)
DNA-Binding Proteins/physiology , Myocarditis , Nitric Oxide Synthase/biosynthesis , Phosphoproteins/physiology , Transcription Factors/physiology , Tyrosine/biosynthesis , Animals , Autoimmunity , Gene Expression Regulation , Humans , Immunohistochemistry , Inflammation/genetics , Inflammation/immunology , Interferon Regulatory Factor-1 , Macrophages/metabolism , Mice , Mice, Knockout , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/physiopathology , Myocardium/metabolism
15.
Immunity ; 6(6): 681-9, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9208841

ABSTRACT

The transcription factor interferon regulatory factor-1 (IRF-1) mediates the effects of IFN. No information exists on its role in lymphokine production. Protection against the intracellular pathogen Leishmania major depends on a Th1 response. Here, we show that CD4+ T cells from Leishmania-infected mice lacking one (+/-) or both (-/-) alleles of the IRF-1 gene developed a profound, gene dose-dependent decrease in IFNgamma production. IRF-1(-/-) mice showed dramatically exacerbated Leishmaniasis. They produced increased Leishmania-specific IgG1 and IgE, and their CD4+ T cells produced increased IL-4, characteristics of the non-protective Th2 response. In cell transfer experiments, IRF-1(-/-) CD4+ T cells mounted normal Th1 responses. However, the ability of IRF-1(-/-) mice to produce IL-12 was severely compromised. Thus, IRF-1 is a determining factor for Th1 responses.


Subject(s)
DNA-Binding Proteins/physiology , Phosphoproteins/physiology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cytokines/biosynthesis , Immunity, Cellular , Immunity, Innate/immunology , Interferon Regulatory Factor-1 , Interleukin-12/biosynthesis , Leishmania major , Leishmaniasis, Cutaneous/immunology , Lymph Nodes/cytology , Lymphocyte Cooperation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/cytology , Th2 Cells/immunology
16.
J Exp Med ; 185(3): 499-505, 1997 Feb 03.
Article in English | MEDLINE | ID: mdl-9053450

ABSTRACT

The interleukin-2 receptor beta chain (IL-2R beta) is expressed on a variety of hematopoietic cell types, including natural killer (NK) cells and nonconventional T lymphocyte subsets such as intestinal intraepithelial lymphocytes (IEL). However, the importance of IL-2R beta-mediated signaling in the growth and development of these cells has yet to be clearly established. We have investigated IEL and NK cells in mice deficient for IL-2R beta and describe here striking defects in the development of these cells. IL-2R beta-/- mice exhibited an abnormal IEL cell population, characterized by a dramatic reduction in T cell receptor alpha beta CD8 alpha alpha and T cell receptor gamma delta lymphocytes. This selective decrease indicates that IEL can be classified into those whose development and/or differentiation is dependent on IL-2R beta function and those for which IL-2R beta-mediated signaling is not essential. NK cell development was also found to be disrupted in IL-2R beta-deficient mice, characterized by a reduction in NK1.1+CD3- cells in the peripheral circulation and an absence of NK cytotoxic activity in vitro. The dependence of NK cells and certain subclasses of IEL cells on IL-2R beta expression points to an essential role for signaling through this receptor, presumably by IL-2 and/or IL-15, in the development of lymphocyte-subsets of extrathymic origin.


Subject(s)
Intestines/immunology , Killer Cells, Natural/physiology , Lymphocytes/physiology , Receptors, Interleukin-2/physiology , Animals , Cytotoxicity, Immunologic , Interleukin-15/physiology , Interleukin-2/physiology , Mice , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, gamma-delta/analysis , Receptors, Interleukin-2/deficiency
17.
J Exp Med ; 184(5): 2043-8, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8920893

ABSTRACT

The activation of natural killer (NK) cells, cytotoxic lymphocytes capable of major histocompatibility complex (MHC)-unrestricted killing and early antiviral defense, is temporally related to the increased interferon (IFN)-alpha/beta production that is seen in the viral infection of mice. Type I IFN (IFN-alpha/beta) are expressed in many cell types early after primary viral infection and have been shown to mediate resistance against a variety of viruses. In this study, the role of the transcriptional activator IFN regulatory factor-1 (IRF-1) in murine NK cell activity was assessed. IRF-1-deficient mice displayed a normal frequency of NK marker-positive cells, but exhibited greatly reduced NK cell-mediated cytotoxicity after both virus infection and stimulation with the IFN inducer polyinosinic:polycytidilic acid in vivo. In vitro, cytolytic activity in IRF-1-deficient NK cells remained defective after stimulation with IFN-beta, IL-2, and IL-12. IRF-1-deficient mice were unable to eliminate syngeneic MHC class I-negative tumor cells in vivo, and had a reduced ability to reject parental semi-allogeneic donor cells from the circulation. Thus, IRF-1 is essential for the induction of NK cell-mediated cytotoxicity and for the in vivo effector functions that are mediated by this activity.


Subject(s)
Cytotoxicity, Immunologic , DNA-Binding Proteins/metabolism , Killer Cells, Natural/immunology , Phosphoproteins/metabolism , Transcription Factors/metabolism , Animals , Antigens/analysis , Antigens, Surface , Cell Transplantation , Cytotoxicity, Immunologic/drug effects , DNA-Binding Proteins/genetics , H-2 Antigens/immunology , Interferon Regulatory Factor-1 , Interferon-beta/pharmacology , Interleukin-12/pharmacology , Interleukin-2/pharmacology , Lectins, C-Type , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , NK Cell Lectin-Like Receptor Subfamily B , Neoplasms, Experimental/immunology , Phosphoproteins/genetics , Proteins/analysis , Transcription Factors/genetics
18.
Science ; 272(5265): 1170-3, 1996 May 24.
Article in English | MEDLINE | ID: mdl-8638161

ABSTRACT

The absence of CTLA-4 results in uncontrolled T cell proliferation. The T cell receptor-specific kinases FYN, LCK, and ZAP-70 as well as the RAS pathway were found to be activated in T cells of Ctla-4-/- mutant mice. In addition, CTLA-4 specifically associated with the tyrosine phosphatase SYP, an interaction mediated by the SRC homology 2 (SH2) domains of SYP and the phosphotyrosine sequence Tyr-Val-Lys-Met within the CTLA-4 cytoplasmic tail. The CTLA-4-associated SYP had phosphatase activity toward the RAS regulator p52SHC. Thus, the RAS pathway and T cell activation through the T cell receptor are regulated by CTLA-4-associated SYP.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Antigens, Differentiation/metabolism , Immunoconjugates , Protein Tyrosine Phosphatases/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Abatacept , Amino Acid Sequence , Animals , Antigens, CD , Antigens, Differentiation/chemistry , CD3 Complex/metabolism , CTLA-4 Antigen , GRB2 Adaptor Protein , Intracellular Signaling Peptides and Proteins , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Recombinant Fusion Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , T-Lymphocytes/immunology , ras Proteins/metabolism , src Homology Domains
19.
Nucl Med Commun ; 17(2): 132-9, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8778637

ABSTRACT

Two 99Tcm-labelled analogues of the chemotactic peptide ForMLF were evaluated as potential agents for imaging inflammation and infection, in the hope that they would be simple to use and would give diagnostically useful images shortly after injection. The peptides differed in the chelation site for 99Tcm and the presence of a hydrophilic spacer. The sequences of RP050 and RP056 were ForNleLFNleYK(G)G-C(Acm)-GPic and ForNleLFNleYKK(DG)GC(Acm)SPic respectively, where Pic is picolinic acid. In in vitro tests of binding to the ForMLF receptor on polymorphonuclear neutrophils and potency for release of myeloperoxidase, RP056 was similar in potency to ForMLF, whereas RP050 was 10 times more potent. When administered in 5-nmol doses to rats, RP050 produced less extensive neutropenia than ForMLF, whereas RP056 produced very little neutropenia. Following labelling by ligand exchange from tartrate or glucoheptonate at 100 degrees C and purification using a C-18 solid-phase extraction cartridge, 4-MBq doses were administered to rats bearing infectious (Escherichia coli) or sterile (zymosan) inflammation sites in the thigh. The inflammation-to-normal muscle ratios at 30 min after injection were 3.9 +/- 0.4 for RP050 and 4.7 +/- 0.3 for RP056 (mean +/- S.E.M., n = 4), and the ratios were maintained for up to 3 h. These peptides are promising agents for imaging inflammation and infection.


Subject(s)
Chemotactic Factors , Inflammation/diagnostic imaging , Oligopeptides , Technetium , Amino Acid Sequence , Animals , Chelating Agents/adverse effects , Chelating Agents/chemistry , Chemotactic Factors/adverse effects , Chemotactic Factors/chemistry , In Vitro Techniques , Male , Molecular Sequence Data , N-Formylmethionine Leucyl-Phenylalanine/analogs & derivatives , N-Formylmethionine Leucyl-Phenylalanine/chemistry , Neutropenia/etiology , Neutropenia/prevention & control , Oligopeptides/adverse effects , Oligopeptides/chemistry , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Receptors, Formyl Peptide , Receptors, Immunologic/metabolism , Receptors, Peptide/metabolism , Technetium/adverse effects
20.
Int Arch Allergy Immunol ; 106(1): 20-4, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7812161

ABSTRACT

Successful induction of the experimental autoimmune disease allergic encephalomyelitis (EAE) depends, in part, upon species susceptibility. The Lewis rat is highly susceptible to EAE whereas the Brown Norway (BN) strain is resistant to induction. Endogenous glucocorticoids influence the manifestation of the disease and recovery from neurological deficits. Moreover, abrogation of the curative steroid-mediated effects converts the condition to a terminal state. In the present study treatment of EAE-inoculated BN rats with the steroid antagonist RU486 (Mifepristone) failed to influence the resistance to symptoms. Similarly, adrenalectomy (ADX) prior to sensitisation did not allow the development of clinical EAE but did facilitate neuroperivascular accumulation of inflammatory-type cells. However, RU486 treatment after ADX induced neurological and histological signs of EAE in the majority of animals. Lymphocyte proliferation studies on cells isolated from BN rats treated with RU486 revealed an enhanced responsiveness to mitogenic and antigenic stimulation. These results strongly implicate endogenous steroids in the expansion of immune cell numbers which would be an absolute requirement for the expression of autoimmune-based neurological disease in otherwise resistant rats.


Subject(s)
Adrenal Cortex Hormones/blood , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Adrenal Cortex Hormones/antagonists & inhibitors , Adrenalectomy , Animals , Cell Division , Lymphocytes/pathology , Male , Mifepristone/pharmacology , Rats , Rats, Inbred BN , Rats, Inbred Lew , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...