Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 281(6): H2549-58, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11709422

ABSTRACT

Early chemokine induction in the area at risk of an ischemic-reperfused (I/R) myocardium is first seen in the venular endothelium. Reperfusion is associated with several induction mechanisms including increased extracellular tumor necrosis factor (TNF)-alpha, reactive oxygen intermediate (ROI) species formation, and adhesion of leukocytes to the venular endothelium. To test the hypothesis that chemokine induction in cardiac venules can occur by ROIs in a TNF-alpha-independent manner, and in the absence of leukocyte accumulation, we utilized wild-type (WT) and TNF-alpha double-receptor knockout mice (DKO) in a closed-chest mouse model of myocardial ischemia (15 min) and reperfusion (3 h), in which there is no infarction. We demonstrate that a single brief period of I/R induces significant upregulation of the chemokines macrophage inflammatory protein (MIP) -1 alpha, -1 beta, and -2 at both the mRNA and protein levels. This induction was independent of TNF-alpha, whereas levels of these chemokines were increased in both WT and DKO mice. Chemokine induction was seen predominantly in the endothelium of small veins and was accompanied by nuclear translocation of nuclear factor-kappa B and c-Jun (AP-1) in venular endothelium. Intravenous infusion of the oxygen radical scavenger N-2-mercaptopropionyl glycine (MPG) initiated 15 min before ischemia and maintained throughout reperfusion obviated chemokine induction, but MPG administration after reperfusion had begun had no effect. The results suggest that ROI generation in the reperfused myocardium rapidly induces C-C and C-X-C chemokines in the venular endothelium in the absence of infarction or irreversible cellular injury.


Subject(s)
Macrophage Inflammatory Proteins/metabolism , Myocardium/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Chemokine CCL3 , Chemokine CCL4 , Chemokine CXCL2 , Endothelium/metabolism , Female , Gene Expression/physiology , Macrophage Inflammatory Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monokines/genetics , Monokines/metabolism , Myocardial Reperfusion Injury/physiopathology , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/genetics
2.
J Exp Med ; 194(4): 519-27, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11514607

ABSTRACT

Bacterial pneumonia is an increasing complication of HIV infection and inversely correlates with the CD4(+) lymphocyte count. Interleukin (IL)-17 is a cytokine produced principally by CD4(+) T cells, which induces granulopoiesis via granulocyte colony-stimulating factor (G-CSF) production and induces CXC chemokines. We hypothesized that IL-17 receptor (IL-17R) signaling is critical for G-CSF and CXC chemokine production and lung host defenses. To test this, we used a model of Klebsiella pneumoniae lung infection in mice genetically deficient in IL-17R or in mice overexpressing a soluble IL-17R. IL-17R-deficient mice were exquisitely sensitive to intranasal K. pneumoniae with 100% mortality after 48 h compared with only 40% mortality in controls. IL-17R knockout (KO) mice displayed a significant delay in neutrophil recruitment into the alveolar space, and had greater dissemination of K. pneumoniae compared with control mice. This defect was associated with a significant reduction in steady-state levels of G-CSF and macrophage inflammatory protein (MIP)-2 mRNA and protein in the lung in response to the K. pneumoniae challenge in IL-17R KO mice. Thus, IL-17R signaling is critical for optimal production of G-CSF and MIP-2 and local control of pulmonary K. pneumoniae infection. These data support impaired IL-17R signaling as a potential mechanism by which deficiency of CD4 lymphocytes predisposes to bacterial pneumonia.


Subject(s)
Chemokines, CXC/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Lung/metabolism , Neutrophils/cytology , Receptors, Interleukin/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Animals , Bronchoalveolar Lavage Fluid , Klebsiella Infections/immunology , Klebsiella pneumoniae/isolation & purification , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin-17 , Recombinant Proteins/genetics
3.
J Immunol ; 167(1): 123-31, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418640

ABSTRACT

CD44 is a widely expressed integral membrane glycoprotein that serves as a specific adhesion receptor for the extracellular matrix glycosaminoglycan hyaluronan. CD44 participates in a variety of physiological and pathological processes through its role in cell adhesion. Under appropriate conditions, the ectodomain of CD44 is proteolytically removed from the cell surface. In this study we show that excessive CD44 shedding can be induced in mouse fibroblasts and monocytes upon exposure of these cells to a CD44-specific Ab immobilized on plastic, whereas treatment with phorbol ester induces significantly enhanced CD44 release from the monocytes only. CD44 shedding proceeds normally in fibroblasts and monocytes deficient in TNF-alpha converting enzyme (TACE), a sheddase involved in the processing of several substrates. Conversely, activation of the CD44 protease has no effect on the release of TNF-alpha from TACE-expressing cells, although the same metalloprotease inhibitor effectively blocks both TACE and the CD44 sheddase. Concomitant with anti-CD44 Ab- or phorbol ester-induced CD44 shedding, dramatic changes are observed in cell morphology and the structure of the actin cytoskeleton. Disruption of actin assembly with cytochalasin reduces CD44 shedding, but not the release of TNF-alpha. Moreover, pharmacological activation of Rho family GTPases Rac1 and Cdc42, which regulate actin filament assembly into distinct cytoskeletal structures, has a profound effect on CD44 release. We conclude that the CD44 sheddase and TACE are distinct enzymes, and that Ab- and phorbol ester-enhanced cleavage of CD44 is controlled in a cell type-dependent fashion by Rho GTPases through the cytoskeleton.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cytoskeleton/immunology , Cytoskeleton/metabolism , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Antibodies, Monoclonal/metabolism , Bone Marrow Cells/immunology , Cell Adhesion/immunology , Cell Line, Transformed , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Culture Media, Conditioned , Fibroblasts/enzymology , Fibroblasts/immunology , Fibroblasts/metabolism , Genes, myc/immunology , Genes, ras/immunology , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/genetics , Hydrolysis , Kinetics , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/deficiency , Mice , Mice, Inbred BALB C , Monocytes/enzymology , Monocytes/immunology , Monocytes/metabolism , RNA, Messenger/biosynthesis , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Tetradecanoylphorbol Acetate/pharmacology
4.
Dev Biol ; 232(1): 204-18, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11254358

ABSTRACT

Many membrane-bound protein precursors, including cytokines and growth factors, are proteolytically shed to yield soluble intercellular regulatory ligands. The responsible protease, tumor necrosis factor-alpha converting enzyme (TACE/ADAM-17), is a transmembrane metalloprotease-disintegrin that cleaves multiple cell surface proteins, although it was initially identified for the enzymatic release of tumor necrosis factor-alpha (TNF-alpha). Mammalian lung growth and development are tightly controlled by cytokines and peptide growth factors. However, the biological function of the cell shedding mechanism during lung organogenesis is not understood. We therefore evaluated the role of TACE as a "sheddase" during lung morphogenesis by analyzing the developmental phenotypes of lungs in mice with an inactive TACE gene in both in vivo and ex vivo organ explant culture. Neonatal TACE-deficient mice had visible respiratory distress and their lungs failed to form normal saccular structures. These newborn mutant lungs had fewer peripheral epithelial sacs with deficient septation and thick-walled mesenchyme, resulting in reduced surface for gas exchange. At the canalicular stage of E16.5, the lungs of TACE mutant mice were impaired in branching morphogenesis, inhibited in epithelial cell proliferation and differentiation, and delayed in vasculogenesis. Embryonic TACE knockout mouse lungs (E12) branched poorly compared to wild-type lungs, when placed into serumless organ culture. Gene expression of both surfactant protein-C and aquaporin-5 were inhibited in cultured TACE-mutant embryonic lungs, indicating defects in both branching and peripheral epithelial cytodifferentiation in the absence of TACE protein. Furthermore, both the hypoplastic phenotype and the delayed cytodifferentiation in TACE-deficient lungs were rescued by exogenous addition of soluble stimulatory factors including either TNF-alpha or epidermal growth factor in embryonic lung culture. Thus, the impaired lung branching and maturation without TACE suggest a broad role for TACE in the processing of multiple membrane-anchored proteins, one or more of which is essential for normal lung morphogenesis. Taken together, our data indicate that the TACE-mediated proteolytic mechanism which enzymatically releases membrane-tethered proteins plays an indispensable role in lung morphogenesis, and its inactivation leads to abnormal lung development.


Subject(s)
Lung/embryology , Membrane Proteins/metabolism , Metalloendopeptidases/physiology , Tumor Necrosis Factor-alpha/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Cell Differentiation , Cell Division , Epidermal Growth Factor/pharmacology , Epithelial Cells/physiology , Lung/pathology , Metalloendopeptidases/genetics , Mice , Mice, Inbred DBA , Mice, Knockout , Morphogenesis , RNA, Messenger/analysis
5.
J Immunol ; 166(5): 3174-83, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11207270

ABSTRACT

4-1BB is expressed on activated CD4(+) and CD8(+) T cells; its ligand, 4-1BB ligand is expressed on APCs. Despite expression on both T cell subpopulations, 4-1BB has been reported to predominantly affect CD8(+) T cell responses. By quantifying graft-vs-host disease alloresponses in vivo, we demonstrate that both CD4(+) and CD8(+) T cell-mediated alloresponses are regulated by 4-1BB/4-1BB ligand interactions to approximately the same extent. 4-1BB receptor-facilitated CD4(+) T cell-mediated alloresponses were partly CD28 independent. In two distinct marrow graft rejection systems, host CD8(+) and CD4(+) T cells each separately contributed to host anti-donor T cell-mediated allograft rejection. alpha 4-1BB mAb increased the graft-vs-leukemia effect of a suboptimal number of donor splenocytes given later post bone marrow transplantation by bolstering allogeneic responses resulting in leukemia elimination. In summary, 4-1BB ligation is a potent regulator of CD4(+) and CD8(+) T cell-mediated allogeneic responses in vivo. Modifying the ligation of 4-1BB represents a new approach to altering the graft-vs-host disease and graft-vs-leukemia effects of allogeneic T cells post bone marrow transplantation.


Subject(s)
Bone Marrow Transplantation/immunology , Graft Rejection/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/physiology , 4-1BB Ligand , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD , Bone Marrow Transplantation/mortality , CD28 Antigens/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Graft Rejection/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/mortality , Graft vs Leukemia Effect/genetics , Injections, Intraperitoneal , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/prevention & control , Ligands , Lymphocyte Activation , Lymphocyte Transfusion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Transplantation, Homologous , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
6.
J Immunol ; 166(3): 1583-9, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160199

ABSTRACT

We previously reported that macrophage activators such as LPS, IL-2, and IL-4 down-modulate the M-CSFR via a mechanism involving protein kinase C and phospholipase C. In this study, we showed that M-CSFR is shed from macrophage surface and identified the protease responsible for M-CSFR cleavage and down-modulation. The shedding of M-CSFR elicited by phorbol esters (tetradecanoylphorbol myristate acetate (TPA)) or LPS in murine BAC.1-2F5 macrophages was prevented by cation chelators, as well as hydroxamate-based competitive inhibitors of metalloproteases. We found that the protease cleaving M-CSFR is a transmembrane enzyme and that its expression is controlled by furin-like serine endoproteases, which selectively process transmembrane metalloproteases. M-CSFR down-modulation was inhibited by treating cells in vivo, before TPA stimulation, with an Ab raised against the extracellular, catalytic domain of proTNF-converting enzyme (TACE). TACE expression was confirmed in BAC.1-2F5 cells and found inhibited after blocking furin-dependent processing. Using TACE-negative murine Dexter-ras-myc cell monocytes, we found that in these cells TPA is unable to down-modulate M-CSFR expression. These data indicated that TACE is required for the TPA-induced M-CSFR cleavage. The possibility that the cleavage is indirectly driven by TACE via the release of TNF was excluded by treating cells in vivo with anti-TNF Ab. Thus, we concluded that TACE is the protease responsible for M-CSFR shedding and down-modulation in mononuclear phagocytes undergoing activation. The possible physiological relevance of this mechanism is discussed.


Subject(s)
Macrophage Activation/immunology , Macrophages/enzymology , Macrophages/immunology , Metalloendopeptidases/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Antibodies, Monoclonal/pharmacology , Catalytic Domain/immunology , Cell Line , Cell Line, Transformed , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Humans , Hydrolysis , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Metalloendopeptidases/immunology , Metalloendopeptidases/isolation & purification , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/enzymology , Monocytes/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
7.
J Biol Chem ; 275(19): 14608-14, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10799547

ABSTRACT

Many membrane-bound proteins, including cytokines, receptors, and growth factors, are proteolytically cleaved to release a soluble form of their extracellular domain. The tumor necrosis factor (TNF)-alpha converting enzyme (TACE/ADAM-17) is a transmembrane metalloproteinase responsible for the proteolytic release or "shedding" of several cell-surface proteins, including TNF and p75 TNFR. We established a TACE-reconstitution system using TACE-deficient cells co-transfected with TACE and substrate cDNAs to study TACE function and regulation. Using the TACE-reconstitution system, we identified two additional substrates of TACE, interleukin (IL)-1R-II and p55 TNFR. Using truncations and chimeric constructs of TACE and another ADAM family member, ADAM-10, we studied the function of the different domains of TACE in three shedding activities. We found that TACE must be expressed with its membrane-anchoring domain for phorbol ester-stimulated shedding of TNF, p75 TNFR, and IL-1R-II, but that the cytoplasmic domain is not required for the shedding of these substrates. The catalytic domain of ADAM-10 could not be functionally substituted for that of TACE. IL-1R-II shedding required the cysteine-rich domain of TACE as well as the catalytic domain, whereas TNF and p75 TNFR shedding required only the tethered TACE catalytic domain.


Subject(s)
Metalloendopeptidases/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Antigens, CD/metabolism , Catalytic Domain , Cell Line , Cytoplasm/enzymology , DNA, Complementary , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Mice , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type II , Transfection , Tumor Necrosis Factor-alpha/metabolism
8.
Blood ; 95(11): 3489-97, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10828034

ABSTRACT

The ligand for the receptor tyrosine kinase fms-like tyrosine kinase 3 (flt3), also referred to as fetal liver kinase-2 (flk-2), has an important role in hematopoiesis. The flt3 ligand (flt3L) is a growth factor for hematopoietic progenitors and induces hematopoietic progenitor and stem cell mobilization in vivo. In addition, when mice are treated with flt3L immature B cells, natural killer (NK) cells and dendritic cells (DC) are expanded in vivo. To further elucidate the role of flt3L in hematopoiesis, mice lacking flt3L (flt3L-/-) were generated by targeted gene disruption. Leukocyte cellularity was reduced in the bone marrow, peripheral blood, lymph nodes (LN), and spleen. Thymic cellularity, blood hematocrit, and platelet numbers were not affected. Significantly reduced numbers of myeloid and B-lymphoid progenitors were noted in the BM of flt3L-/- mice. In addition a marked deficiency of NK cells in the spleen was noted. DC numbers were also reduced in the spleen, LN, and thymus. Both myeloid-related (CD11c(++) CD8alpha(-)) and lymphoid-related (CD11c(++) CD8alpha(+)) DC numbers were affected. We conclude that flt3L has an important role in the expansion of early hematopoietic progenitors and in the generation of mature peripheral leukocytes.


Subject(s)
B-Lymphocytes/cytology , Dendritic Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/immunology , Killer Cells, Natural/cytology , Membrane Proteins/physiology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bone Marrow/immunology , Colony-Forming Units Assay , Dendritic Cells/drug effects , Dendritic Cells/immunology , Genomic Library , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-7/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Kinetics , Leukocytes/cytology , Ligands , Lymph Nodes/immunology , Lymphocyte Culture Test, Mixed , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/pharmacology , Recombinant Proteins/pharmacology , Spleen/immunology , Thymus Gland/immunology
9.
Cell Immunol ; 201(1): 33-41, 2000 Apr 10.
Article in English | MEDLINE | ID: mdl-10805971

ABSTRACT

The development and function of secondary lymphoid tissue require signaling by tumor necrosis factor and lymphotoxins. Mice deficient in LTbetaR show defective organogenesis of lymph nodes and Peyer's patches and a severely disturbed splenic architecture. In contrast, TNF or p55TNF-R deficiency does not affect the organogenesis of peripheral lymphoid organs but interferes with the formation of B cell follicles and the appearance of FDC networks and germinal centers in all secondary lymphoid organs. Based on these differences, we have previously hypothesized that the role of TNF in lymphoid structure is distinct from that of LT and restricted in regulating cellular interactions that allow the differentiation and/or correct positioning of FDCs. In the present study we show that, in addition to the defects in follicular structure, TNF or p55TNF-R knockout mice exhibit defects in the formation of the macrophage populations and of the sinus lining cells of the splenic marginal zone. Interestingly, a large number of dendritic-shaped cells stained with FDC-specific markers and able to trap immune complexes are retained within the defective marginal zone of TNF and p55TNF-R knockout spleens. We conclude that the primary defect in the lymphoid phenotype of TNF or p55TNF-R knockout mice is the failure of FDC precursors to migrate through the disorganized marginal sinus and to home properly into the splenic follicular areas where they would promote the formation of B cell follicles and germinal centers.


Subject(s)
Antigens, CD/physiology , Cell Movement/physiology , Dendritic Cells, Follicular/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antigens, CD/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Spleen/cytology , Stem Cells/physiology , Tumor Necrosis Factor-alpha/genetics
10.
Proc Natl Acad Sci U S A ; 97(10): 5456-61, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10779546

ABSTRACT

Previous studies have shown that proinflammatory cytokines, such as tumor necrosis factor (TNF), are expressed after acute hemodynamic overloading and myocardial ischemia/infarction. To define the role of TNF in the setting of ischemia/infarction, we performed a series of acute coronary artery occlusions in mice lacking one or both TNF receptors. Left ventricular infarct size was assessed at 24 h after acute coronary occlusion by triphenyltetrazolium chloride (TTC) staining in wild-type (both TNF receptors present) and mice lacking either the type 1 (TNFR1), type 2 (TNFR2), or both TNF receptors (TNFR1/TNFR2). Left ventricular infarct size as assessed by TTC staining was significantly greater (P < 0.005) in the TNFR1/TNFR2-deficient mice (77.2% +/- 15.3%) when compared with either wild-type mice (46.8% +/- 19.4%) or TNFR1-deficient (47.9% +/- 10.6%) or TNFR2-deficient (41.6% +/- 16.5%) mice. Examination of the extent of necrosis in wild-type and TNFR1/TNFR2-deficient mice by anti-myosin Ab staining demonstrated no significant difference between groups; however, the peak frequency and extent of apoptosis were accelerated in the TNFR1/TNFR2-deficient mice when compared with the wild-type mice. The increase in apoptosis in the TNFR1/TNFR2-deficient mice did not appear to be secondary to a selective up-regulation of the Fas ligand/receptor system in these mice. These data suggest that TNF signaling gives rise to one or more cytoprotective signals that prevent and/or delay the development of cardiac myocyte apoptosis after acute ischemic injury.


Subject(s)
Antigens, CD/physiology , Apoptosis , Myocardial Infarction/prevention & control , Myocardial Ischemia/physiopathology , Myocardium/pathology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Acute Disease , Animals , Antigens, CD/genetics , Coronary Vessels/physiology , Coronary Vessels/physiopathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardial Ischemia/pathology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I
11.
J Biol Chem ; 275(14): 10379-87, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10744726

ABSTRACT

HER4 is a member of the epidermal growth factor receptor family and has an essential function in heart and neural development. Identification of two HER4 isoforms, HER4 JM-a and JM-b, which differ in their extracellular juxtamembrane region and in their susceptibility to cleavage after phorbol ester stimulation, showed that the juxtamembrane region of the receptor is critical for proteolysis. We now demonstrate that phorbol ester and pervanadate are effective stimuli for HER4 JM-a processing and that the HER4 JM-b isoform does not undergo cleavage in response to any of the stimuli studied. We also show that HER4 JM-a is not cleaved in cells lacking the metalloprotease tumor necrosis factor-alpha-converting enzyme (TACE) and that reexpression of TACE in these cells restores constitutive and regulated processing of HER4 JM-a. Moreover, we show that the sequence specific to the HER4 JM-a juxtamembrane region is sufficient to confer susceptibility to phorbol 12-myristate 13-acetate-induced cleavage of the HER2 receptor. In conclusion, we provide evidence that TACE is essential for the regulated shedding of the HER4 JM-a receptor.


Subject(s)
Drosophila Proteins , ErbB Receptors/metabolism , Metalloendopeptidases/metabolism , 3T3 Cells , ADAM Proteins , ADAM17 Protein , Animals , Clone Cells , Cloning, Molecular , Colforsin/pharmacology , Disintegrins/metabolism , Kinetics , Marine Toxins , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Protein Isoforms/metabolism , Receptor, ErbB-4 , Recombinant Proteins/metabolism , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Transfection , Tumor Necrosis Factor-alpha/metabolism , Vanadates/pharmacology
12.
J Exp Med ; 191(5): 771-80, 2000 Mar 06.
Article in English | MEDLINE | ID: mdl-10704459

ABSTRACT

C57BL/6 mice genetically deficient in interleukin 15 (IL-15(-/-) mice) were generated by gene targeting. IL-15(-/-) mice displayed marked reductions in numbers of thymic and peripheral natural killer (NK) T cells, memory phenotype CD8(+) T cells, and distinct subpopulations of intestinal intraepithelial lymphocytes (IELs). The reduction but not absence of these populations in IL-15(-/-) mice likely reflects an important role for IL-15 for expansion and/or survival of these cells. IL-15(-/-) mice lacked NK cells, as assessed by both immunophenotyping and functional criteria, indicating an obligate role for IL-15 in the development and functional maturation of NK cells. Specific defects associated with IL-15 deficiency were reversed by in vivo administration of exogenous IL-15. Despite their immunological defects, IL-15(-/-) mice remained healthy when maintained under specific pathogen-free conditions. However, IL-15(-/-) mice are likely to have compromised host defense responses to various pathogens, as they were unable to mount a protective response to challenge with vaccinia virus. These data reveal critical roles for IL-15 in the development of specific lymphoid lineages. Moreover, the ability to rescue lymphoid defects in IL-15(-/-) mice by IL-15 administration represents a powerful means by which to further elucidate the biological roles of this cytokine.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interleukin-15/immunology , Killer Cells, Natural/immunology , Receptors, Interleukin-2/immunology , Animals , Cell Lineage , Epithelial Cells/immunology , Female , Interleukin-15/genetics , Lymph Nodes/anatomy & histology , Lymph Nodes/immunology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Organ Size , Receptors, Interleukin-15 , Receptors, Interleukin-2/genetics , Spleen/anatomy & histology , Spleen/immunology , Thymus Gland/anatomy & histology , Thymus Gland/immunology , Vaccinia/mortality
13.
Am J Respir Cell Mol Biol ; 22(1): 85-91, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10615069

ABSTRACT

We hypothesized that tumor necrosis factor (TNF)-alpha signaling is essential to inflammation and host defense during Escherichia coli pneumonia. We tested this hypothesis by instilling E. coli into the lungs of wild-type (WT) mice and gene-targeted mice that lack both p55 and p75 receptors for TNF-alpha. The emigration of neutrophils 6 h after instillation of E. coli was not decreased, but rather was significantly increased (167% of WT), in TNF receptor (TNFR)-deficient mice. This increased neutrophil emigration did not result from peripheral blood neutrophilia or enhanced neutrophil sequestration, inasmuch as the numbers of neutrophils in the circulating blood and in the pulmonary capillaries did not differ between TNFR-deficient and WT mice. The accumulation of pulmonary edema fluid was not inhibited in TNFR-deficient compared with WT mice. Nuclear factor-kappaB (NF-kappaB) translocation in the lungs was not prevented in TNFR-deficient mice. Thus, signaling pathways independent of TNFRs can mediate the acute inflammatory response during E. coli pneumonia. However, despite this inflammatory response, bacterial clearance was impaired in TNFR-deficient mice (109 +/- 8% versus 51 +/- 14% of the original inoculum viable after 6 h in TNFR-deficient and WT mice, respectively). Increased neutrophil emigration during E. coli pneumonia in TNFR-deficient mice may thus result from an increased bacterial burden in the lungs. During acute E. coli pneumonia, the absence of TNFR signaling compromised bacterial killing, but did not prevent inflammation, as measured by the accumulation of edema fluid and neutrophils.


Subject(s)
Pneumonia, Bacterial/metabolism , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Biological Transport , Inflammation/metabolism , Inflammation/microbiology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/mortality , Pneumonia, Bacterial/pathology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II
14.
J Immunol ; 163(9): 4833-41, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10528184

ABSTRACT

4-1BB ligand (4-1BBL) is a member of the TNF family expressed on activated APC. 4-1BBL binds to 4-1BB (CD137) on activated CD4 and CD8 T cells and in conjunction with strong signals through the TCR provides a CD28-independent costimulatory signal leading to high level IL-2 production by primary resting T cells. Here we report the immunological characterization of mice lacking 4-1BBL and of mice lacking both 4-1BBL and CD28. 4-1BBL-/- mice mount neutralizing IgM and IgG responses to vesicular stomatitis virus that are indistinguishable from those of wild-type mice. 4-1BBL-/- mice show unimpaired CTL responses to lymphocytic choriomeningitis virus (LCMV) and exhibit normal skin allograft rejection but have a weaker CTL response to influenza virus than wild-type mice. 4-1BBL-/-CD28-/- mice retain the CTL response to LCMV, respond poorly to influenza virus, and exhibit a delay in skin allograft rejection. In agreement with these in vivo results, allogeneic CTL responses of CD28-/- but not CD28+/+ T cells to 4-1BBL-expressing APC are substantially inhibited by soluble 4-1BB receptor as is the in vitro secondary response of CD28+ T cells to influenza virus peptides. TCR-transgenic CD28-/- LCMV glycoprotein-specific T cells are insensitive to the presence of 4-1BBL when a wild-type peptide is used, but the response to a weak agonist peptide is greatly augmented by the presence of 4-1BBL. These results further substantiate the idea that different immune responses vary in their dependence on costimulation and suggest a role for 4-1BBL in augmenting suboptimal CTL responses in vivo.


Subject(s)
CD28 Antigens/genetics , Cytotoxicity, Immunologic/genetics , Graft Rejection/immunology , Influenza A virus/immunology , Skin Transplantation/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , 4-1BB Ligand , Animals , Antibodies, Viral/biosynthesis , CD28 Antigens/physiology , Female , Gene Targeting , Graft Rejection/genetics , Ligands , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Cytotoxic/virology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/physiology , Vesicular stomatitis Indiana virus/immunology
15.
Genes Dev ; 13(18): 2412-24, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10500098

ABSTRACT

The physiological role of the TNF receptor (TNFR) family member, RANK, was investigated by generating RANK-deficient mice. RANK(-/-) mice were characterized by profound osteopetrosis resulting from an apparent block in osteoclast differentiation. RANK expression was not required for the commitment, differentiation, and functional maturation of macrophages and dendritic cells from their myeloid precursors but provided a necessary and specific signal for the differentiation of myeloid-derived osteoclasts. RANK(-/-) mice also exhibited a marked deficiency of B cells in the spleen. RANK(-/-) mice retained mucosal-associated lymphoid tissues including Peyer's patches but completely lacked all other peripheral lymph nodes, highlighting an additional major role for RANK in lymph node formation. These experiments reveal that RANK provides critical signals necessary for lymph node organogenesis and osteoclast differentiation.


Subject(s)
Carrier Proteins , Lymph Nodes/embryology , Membrane Glycoproteins , Osteoclasts/physiology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/physiology , Animals , B-Lymphocytes/physiology , Bone Resorption/metabolism , Bone and Bones/diagnostic imaging , Dendritic Cells/physiology , Flow Cytometry , Gene Targeting , Hematopoiesis, Extramedullary/genetics , Hematopoietic Stem Cells/cytology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteopetrosis/diagnostic imaging , Osteopetrosis/metabolism , Peyer's Patches/anatomy & histology , Phenotype , RANK Ligand , Radiography , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor/metabolism , Spleen/anatomy & histology , Spleen/embryology
16.
Ann N Y Acad Sci ; 878: 442-52, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415747

ABSTRACT

The adamalysins are a family of proteins in the metzincin superfamily of metalloproteases, which also includes the matrix metalloproteinases. There are two subfamilies of adamalysins: the snake venom metalloproteases (SVMPs) and the ADAMs (proteins containing a disintegrin and metalloprotease domain). At least 23 ADAMs have been identified to date. The ADAMs are expressed by a wide variety of cell types, and are involved in functions as diverse as sperm-egg binding, myotube formation, neurogenesis, and proteolytic processing of cell surface proteins. An overview of the ADAM family and their functions will be presented. TACE is a unique member of the ADAM family that cleaves membrane-bound TNF-alpha to generate soluble TNF-alpha. Mice lacking proteolytically active TACE have been generated and characterized. The TACE knock-out results in perinatal lethality. Cells from the TACE-deficient mice release 80-90% less soluble TNF-alpha than do wild-type cells. Irradiated mice that are reconstituted with TACE knock-out hematopoeitic stem cells have markedly reduced levels of serum TNF-alpha following LPS challenge, compared to irradiated mice reconstituted with wild-type cells, suggesting that TACE is the major TNF-alpha converting enzyme in vivo. TACE-deficient cells are compromised in the generation of other soluble proteins that are produced as the result of cleavage of a membrane precursor form, suggesting that TACE is involved in multiple shedding events.


Subject(s)
Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , ADAM Proteins , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases , Animals , Female , Humans , Male , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Snake Venoms/metabolism , Sperm-Ovum Interactions , Tumor Necrosis Factor-alpha/metabolism
17.
J Immunol ; 163(2): 920-6, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10395688

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL) is able to kill many transformed cells of diverse tissue types. We show that TRAIL is inducible by IFN-gamma, by TNF-alpha, and by infection with human CMV, and has potent antiviral activity in vitro. CMV infection and IFN-gamma also reciprocally modulate TRAIL receptor (TRAIL-R) expression. CMV infection increased the expression of TRAIL-R1 and -R2, whereas IFN-gamma down-regulated the expression of TRAIL-Rs on uninfected fibroblasts. Moreover, IFN-gamma significantly decreased the basal level of NF-kappaB activation, a known survival factor that inhibits apoptosis. Thus, TRAIL selectively kills virus-infected cells while leaving uninfected cells intact, and IFN-gamma potentiates these effects by dynamic modulation of TRAIL and TRAIL-R expression and by sensitizing cells to apoptosis. The regulation of TRAIL and TRAIL-R expression may represent a general mechanism that contributes to the control of TRAIL-mediated apoptosis.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antiviral Agents/pharmacology , Interferon-gamma/physiology , Membrane Glycoproteins/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Apoptosis Regulatory Proteins , Cell Death/immunology , Cells, Cultured , Cytokines/physiology , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Drug Synergism , Fibroblasts , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , RNA, Messenger/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology , Virus Replication/immunology
18.
Am J Physiol ; 276(5): L715-27, 1999 05.
Article in English | MEDLINE | ID: mdl-10330027

ABSTRACT

To determine the roles of the type 1 tumor necrosis factor (TNF) receptor (TNFR1) in lung inflammation and antibacterial defense, we exposed transgenic mice lacking TNFR1 [TNFR1(-/-)] and wild-type control mice to aerosolized lipopolysaccharide or Pseudomonas aeruginosa. After LPS, bronchoalveolar lavage fluid (BALF) from TNFR1(-/-) mice contained fewer neutrophils and less macrophage inflammatory protein-2 than BALF from control mice. TNF-alpha, interleukin-1beta, and total protein levels in BALF as well as tissue intercellular adhesion molecule-1 expression did not differ between the two groups. In contrast, lung inflammation and bacterial clearance after infection were augmented in TNFR1(-/-) mice. BALF from infected TNFR1(-/-) mice contained more neutrophils and TNF-alpha and less interleukin-1beta and macrophage inflammatory protein-2 than that from control mice, but protein levels were similarly elevated in both groups. Lung inflammation and bacterial clearance were also augmented in mice lacking both TNF receptors. Thus TNFR1 facilitates neutrophil recruitment after inhalation of lipopolysaccharide, in part by augmenting chemokine induction. In contrast, TNFR1 attenuates lung inflammation in response to live bacteria but does not contribute to increased lung permeability and is not required for the elimination of P. aeruginosa.


Subject(s)
Endotoxins/administration & dosage , Pneumonia, Bacterial/microbiology , Pseudomonas Infections , Receptors, Tumor Necrosis Factor/physiology , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2 , Chemotaxis, Leukocyte , Cytokines/analysis , Intercellular Adhesion Molecule-1/analysis , Interleukin-1/analysis , Leukocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monokines/analysis , Neutrophils , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Proteins/analysis , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/analysis
19.
J Immunol ; 162(8): 4943-51, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10202041

ABSTRACT

The mechanism of cytokine-induced shock remains poorly understood. The combination of IL-2 and IL-12 has synergistic antitumor activity in vivo, yet has been associated with significant toxicity. We examined the effects of IL-2 plus IL-12 in a murine model and found that the daily, simultaneous administration of IL-2 and IL-12 resulted in shock and 100% mortality within 4 to 12 days depending on the strain employed. Mice treated with IL-2 plus IL-12 exhibited NK cell apoptosis, pulmonary edema, degenerative lesions of the gastrointestinal tract, and elevated serum levels of proinflammatory cytokines and acute phase reactants. The actions of TNF-alpha, IFN-gamma, macrophage-inflammatory protein-1alpha, IL-1, IL-1-converting enzyme, Fas, perforin, inducible nitric oxide synthase, and STAT1 did not contribute to the observed toxicity, nor did B or T cells. However, toxicity and death from treatment with IL-2 plus IL-12 could be completely abrogated by elimination of NK cells. These results suggest that the fatal systemic inflammatory response induced by this cytokine treatment is critically dependent upon NK cells, but does not appear to be mediated by the known effector molecules of this cellular compartment. These data may provide insight into the pathogenesis of cytokine-induced shock in humans.


Subject(s)
Interleukins/adverse effects , Killer Cells, Natural/immunology , Shock, Septic/etiology , Shock, Septic/mortality , Animals , Cell Separation , Cytokines/biosynthesis , Cytokines/blood , Drug Therapy, Combination , Female , Interferon-gamma/physiology , Interleukin-1/physiology , Interleukin-12/administration & dosage , Interleukin-12/adverse effects , Interleukin-15/administration & dosage , Interleukin-15/adverse effects , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Interleukins/administration & dosage , Liver/pathology , Lung/pathology , Macrophage Activation , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Mice, Transgenic , Monocytes/immunology , Shock, Septic/immunology , Shock, Septic/pathology , Spleen/pathology , Tumor Necrosis Factor-alpha/physiology
20.
Am J Respir Cell Mol Biol ; 20(4): 825-33, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10101016

ABSTRACT

We have investigated a potential role for tumor necrosis factor (TNF)-alpha and its two receptors (p55 and p75) in lung injury. We used several varieties of mice exposed endotracheally to two fibrogenic agents, silica (0.2 g/kg) and bleomycin (4 U/kg). The lungs were analyzed at 14 and 28 d after exposure to bleomycin or silica, respectively, for TNF and TNF receptor (TNFR) messenger RNA (mRNA), hydroxyproline content, and histopathology. Silica induced increased (over saline-treated animals) expression of TNF mRNA in double TNFR knockout (Ko), C57BL/6, BALB/c, and 129/J mice. In contrast, bleomycin increased expression in all but BALB/c mice, which are resistant to the fibrogenic effects of this drug. mRNA expression of both receptors was constitutively expressed in all of the normal murine strains. Silica upregulated expression of the p75 receptor, but not the p55 receptor, in the C57BL/6, BALB/c, and 129/J mice. In comparison, bleomycin had little effect on either receptor in the bleomycin-resistant BALB/c mice. Hydroxyproline content of the lungs after treatment followed this same pattern, with significant increases caused by silica in the C57BL/6, BALB/c, and 129/J mice, whereas bleomycin caused no apparent increases in the BALB/c mice. Even though silica and bleomycin induced increases in TNF in the TNFR Ko mice, the mice were protected from the fibrogenic effects of these agents. This study supports the concept that TNF is a central mediator of interstitial pulmonary fibrosis.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Bleomycin/toxicity , Lung/pathology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Silicon Dioxide/toxicity , Transcription, Genetic/drug effects , Animals , Antigens, CD/physiology , Crosses, Genetic , Female , Gene Expression Regulation/drug effects , Lung/drug effects , Lung/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional , RNA, Messenger/genetics , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , Recombination, Genetic , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...