Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 449
Filter
Add more filters

Publication year range
1.
Clin Exp Immunol ; 172(3): 500-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23600839

ABSTRACT

Recent basic and clinical studies have shown that the programmed death ligand (PD-L)/PD-1 pathway has a significant role in tumour immunity, and its blockade has a therapeutic potential against several human cancers. We hypothesized that anti-angiogeneic treatment might augment the efficacy of PD-1 blockade. To this end, we evaluated combining the blockade of PD-1 and vascular endothelial growth factor receptor 2 (VEGFR2) in a murine cancer model using Colon-26 adenocarcinoma. Interestingly, simultaneous treatment with anti-PD-1 and anti-VEGFR2 monoclonal antibodies (mAbs) inhibited tumour growth synergistically in vivo without overt toxicity. Blocking VEGFR2 inhibited tumour neovascularization significantly, as demonstrated by the reduced number of microvessels, while PD-1 blockade had no impact on tumour angiogenesis. PD-1 blockade might promote T cell infiltration into tumours and significantly enhanced local immune activation, as shown by the up-regulation of several proinflammatory cytokine expressions. Importantly, VEGFR2 blockade did not interfere with T cell infiltration and immunological activation induced by PD-1 blockade. In conclusion, simultaneous blockade of PD-1 and VEGFR2 induced a synergistic in-vivo anti-tumour effect, possibly through different mechanisms that might not be mutually exclusive. This unique therapeutic strategy may hold significant promise for future clinical application.


Subject(s)
Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenocarcinoma/blood supply , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Cell Line, Tumor , Colonic Neoplasms/blood supply , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Drug Synergism , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/immunology , Neovascularization, Pathologic/prevention & control , Programmed Cell Death 1 Receptor/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology
2.
Gene Ther ; 19(11): 1075-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22071968

ABSTRACT

Myeloablative transplantation of bone marrow (BM) engineered to express myelin oligodendrocyte glycoprotein (MOG) establishes central intrathymic tolerance and completely prevents MOG-induced experimental autoimmune encephalomyelitis (EAE) in mice. Here we asked whether non-myeloablative transplantation of MOG expressing BM (pMOG-bone marrow transplantation (BMT)) can also provide the same protection. Using stepwise reduction of irradiation doses, 275 cGy irradiation with pMOG-BMT protected 100% of mice from EAE development even with two subsequent re-challenge with MOG. Irradiation doses <275 cGy produced dose-dependent partial protection with significant disease protection still evident at 50 cGy. Splenocytes from 275 cGy recipients proliferated to MOG stimulation in vitro, indicating that MOG-reactive cells are present in the periphery but failed to induce disease. MOG-stimulated splenocytes produced little or no interleukin-17, interferon-ƎĀ³, granulocyte-monocyte colony stimulating factor and tumor necrosis factor-α compared with EAE control. Adoptive transfer of CD4 T cells from EAE-resistant mice into Rag2(-/-) mice devoid of MOG expression resulted in MOG-induced EAE in ~74% of mice. Treatment of EAE-resistant mice with anti-programmed death 1 (PD-1) monoclonal antibody-induced EAE in 67% of mice. We conclude that non-myeloablative transplantation of self-antigen expressing BM induces robust peripheral tolerance that completely prevented EAE development. Our findings implicate clonal anergy and the PD-1 pathway in the maintenance of peripheral tolerance.


Subject(s)
Autoantigens/genetics , Autoantigens/immunology , Autoimmunity/genetics , Bone Marrow Transplantation , Peripheral Tolerance/genetics , Transplantation Conditioning , Adoptive Transfer , Animals , Antibodies/administration & dosage , Antibodies/immunology , Antigens, Differentiation/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Cytokines/immunology , Dose-Response Relationship, Radiation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Inflammation Mediators/immunology , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein/immunology , Programmed Cell Death 1 Receptor , Spinal Cord/pathology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Regulatory/immunology , Transplantation Chimera , Whole-Body Irradiation
3.
Allergy ; 67(3): 328-35, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22175699

ABSTRACT

BACKGROUND: Eosinophils and lymphocytes are pathogenically important in allergic inflammation and sensitive to Fas-mediated apoptosis. Fas ligand (FasL) activity therefore should play a role in regulating the allergic immune response. We aimed to characterize the role of FasL expression in airway eosinophilia in Aspergillus fumigatus (Af)-induced sensitization and to determine whether FasL neutralization alters the inflammatory response. METHODS: Sensitized Balb/c mice were killed before (day 0) and 1, 7 and 10 days after a single intranasal challenge with Af. Animals received either neutralizing antibody to FasL (clone MFL4) or irrelevant hamster IgG via intraperitoneal injection on days -1 and 5. FasL expression, BAL and tissue inflammatory cell and cytokine profile, and apoptosis were assessed. RESULTS: Postchallenge FasL gene expression in BAL cells and TUNEL positivity in the airways coincided with the height of inflammatory cell influx on day 1, while soluble FasL protein was released on day 7, preceding resolution of the inflammatory changes. Although eosinophil numbers showed a negative correlation with soluble FasL levels in the airways, MBP(+) eosinophils remained TUNEL negative in the submucosal tissue, throughout the 10-day period after Af challenge. Systemic FasL neutralization significantly enhanced BAL and tissue eosinophil counts. This effect was associated with increased activation of T cells and release of IL-5, IL-9, and GM-CSF in the BAL fluid of mice, indicating an involvement of pro-eosinophilic survival pathways. CONCLUSIONS: FasL activity may play an active role in resolving eosinophilic inflammation through regulating T cells and pro-eosinophilic cytokine release during the allergic airway response.


Subject(s)
Asthma/immunology , Asthma/physiopathology , Disease Models, Animal , Eosinophils/immunology , Fas Ligand Protein/immunology , Inflammation/immunology , Animals , Aspergillus fumigatus/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cricetinae , Eosinophilia/immunology , Eosinophilia/pathology , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Humans , Leukocyte Count , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred BALB C , Neutralization Tests
4.
Nat Med ; 3(2): 165-70, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9018234

ABSTRACT

Activation of the cell-surface antigen CD95 induces apoptosis of CD95-bearing tumor cells. In this study, we investigated the antitumor effect of locally produced CD95 ligand (CD95L) on CD95-negative tumor cells in vivo. Introduction of CD95L cDNA into murine tumor cells did not affect growth in vitro but caused rejection in vivo. Neutrophils were primarily responsible for this rejection. A CD8 T cell-mediated protective immunity against subsequent challenge with parental tumor cells was also elicited. These results provide evidence for the potential utility of CD95L in tumor eradication and also reveal a proinflammatory function of CD95L.


Subject(s)
Apoptosis/immunology , Immunity, Cellular , Membrane Glycoproteins/immunology , Neoplasms, Experimental/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , Fas Ligand Protein , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Humans , Membrane Glycoproteins/genetics , Mice , Neoplasms, Experimental/immunology
5.
Nat Med ; 5(8): 930-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10426318

ABSTRACT

Dendritic cells (DCs) are special subsets of antigen-presenting cells characterized by their highly potent capacity to activate immunologically naive T cells. Here we report that DCs that are transfected with CD95 ligand (CD95L) cDNA, called 'killer' DCs, deliver death signals, instead of activation signals, to T cells after antigen-specific interaction. Injection of antigen-pulsed killer DCs into mice before sensitization induced antigen-specific immunosuppression. When administered after sensitization, killer DCs suppressed immune responses almost completely after subsequent challenge. Thus, killer DCs represent an entirely new immunomodulatory protocol, which may become directly applicable in preventing and even treating T cell-mediated inflammatory diseases.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Transplantation , Clone Cells/immunology , DNA, Complementary/genetics , Dendritic Cells/cytology , Dermatitis, Allergic Contact/immunology , Dinitrofluorobenzene , Fas Ligand Protein , Female , Hypersensitivity, Delayed/immunology , Immunity, Cellular/genetics , Liver/drug effects , Mice , Ovalbumin/immunology , Transfection , fas Receptor/immunology
6.
Nat Med ; 7(1): 94-100, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135622

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in various tumor cells in vitro, but its physiological role in tumor surveillance remains unknown. Here, we report that TRAIL is constitutively expressed on murine natural killer (NK) cells in the liver and plays a substantial role in suppressing tumor metastasis. Freshly isolated NK cells, but not natural killer T cells or ordinary T cells, from the liver expressed cell surface TRAIL, which was responsible for spontaneous cytotoxicity against TRAIL-sensitive tumor cells in vitro along with perforin and Fas ligand (FasL). Administration of neutralizing monoclonal antibody against TRAIL significantly increased experimental liver metastases of several TRAIL-sensitive tumor cell lines. Such an anti-metastatic effect of TRAIL was not observed in NK cell-depleted mice or interferon-gamma-deficient mice, the latter of which lacked TRAIL on liver NK cells. These findings provide the first evidence for the physiological function of TRAIL as a tumor suppressor.


Subject(s)
Apoptosis/physiology , Killer Cells, Natural/immunology , Liver/immunology , Membrane Glycoproteins/physiology , Neoplasm Metastasis/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , Ligands , Liver/cytology , Liver Neoplasms, Experimental/secondary , Male , Mice , Mice, Inbred Strains , TNF-Related Apoptosis-Inducing Ligand
7.
J Exp Med ; 170(5): 1757-61, 1989 Nov 01.
Article in English | MEDLINE | ID: mdl-2478655

ABSTRACT

The CD56 differentiation antigen, recognized by anti-Leu-19 and NKH-1 mAbs, is a 200-220-kD glycoprotein that is expressed predominantly on human NK cells and a minor subset of T lymphocytes mediating MHC-unrestricted cytotoxicity. The recent finding that CD56 is an isoform of the neural cell adhesion molecule (N-CAM) prompted us to examine the adhesive function of CD56 in the NK-target cell interaction. Synergistic inhibitory effects of anti-CD56 mAbs with anti-LFA-1 and/or anti-LFA-3 mAbs were demonstrated on NK cell-mediated cytotoxicity and on NK cell binding to target cells only when the target cells also express CD56. These findings indicate that CD56 on NK cells can serve as the third pathway of cell adhesion other than those mediated by the CD2/LFA-3 and LFA-1/ICAM-1 interactions, and is involved in NK cell cytotoxicity when interacting with the cells bearing N-CAM.


Subject(s)
Antigens, CD/physiology , Antigens, Differentiation, T-Lymphocyte/physiology , Cell Adhesion Molecules/physiology , Killer Cells, Natural/physiology , Antibodies, Monoclonal/immunology , Antigens, Differentiation/physiology , Antigens, Surface/physiology , CD56 Antigen , CD58 Antigens , Cytotoxicity, Immunologic , Humans , Immunity, Cellular , Lymphocyte Function-Associated Antigen-1 , Membrane Glycoproteins/physiology , Receptors, Leukocyte-Adhesion/physiology
8.
J Exp Med ; 192(9): 1373-80, 2000 Nov 06.
Article in English | MEDLINE | ID: mdl-11067885

ABSTRACT

TWEAK, a new member of the tumor necrosis factor (TNF) family, induces cell death in some tumor cell lines, but its physiological functions are largely unknown. In this study, we investigated the expression and function of TWEAK in human peripheral blood mononuclear cells (PBMCs) by using newly generated anti-human TWEAK mAbs. Although freshly isolated PBMCs expressed no detectable level of TWEAK on their surfaces, a remarkable TWEAK expression was rapidly observed on monocytes upon stimulation with interferon (IFN)-gamma but not with IFN-alpha or lipopolysaccharide. Cytotoxic activity of IFN-gamma-stimulated monocytes against human squamous carcinoma cell line HSC3 was inhibited partially by anti-TWEAK mAb alone and almost completely by combination with anti-TRAIL (TNF-related apoptosis-inducing ligand) mAb. These results revealed a novel pathway of monocyte cytotoxicity against tumor cells that is mediated by TWEAK and potentiated by IFN-gamma.


Subject(s)
Carrier Proteins/immunology , Cytotoxicity, Immunologic/drug effects , Interferon-gamma/pharmacology , Monocytes/drug effects , Monocytes/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/analysis , Apoptosis Regulatory Proteins , Carcinoma, Squamous Cell/immunology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cells, Cultured , Cytokine TWEAK , Cytotoxicity Tests, Immunologic , Flow Cytometry , Humans , Interferon-alpha/pharmacology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/physiology , Monocytes/metabolism , Recombinant Fusion Proteins , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology , Tumor Necrosis Factors
9.
J Exp Med ; 179(1): 341-6, 1994 Jan 01.
Article in English | MEDLINE | ID: mdl-7903681

ABSTRACT

Indefinite graft survival was obtained with murine cardiac allografts using the combined administration of monoclonal antibodies (mAbs) directed against the receptor ligand pair CD2-CD48. Although each antibody could prolong graft survival when given alone, neither resulted in the indefinite graft survival seen with the combination. Combined mAb administration is associated with inhibition of T cell priming and help and subsequent cytotoxic T lymphocyte generation. This indicates that the interaction between CD2 and its ligand is important for antigen priming and recognition, and combined mAbs may prove to be a useful therapeutic regimen for transplantation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Graft Survival/immunology , Heart Transplantation/immunology , Receptors, Immunologic/immunology , Animals , CD2 Antigens , CD48 Antigen , Drug Synergism , Female , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Homologous
10.
J Exp Med ; 189(9): 1451-60, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10224285

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a proapoptotic member of the TNF family of type II membrane proteins, which constitutes one component of T cell cytotoxicity. In this study, we investigated the expression and function of TRAIL in human peripheral blood T (PBT) cells. Although freshly isolated PBT cells did not express a detectable level of TRAIL on their surface, a remarkable TRAIL expression was rapidly induced on the surface of both CD4(+) and CD8(+) PBT cells upon stimulation with anti-CD3 monoclonal antibody and type I interferons (IFNs). This enhancement of TRAIL expression was a unique feature of type I IFNs (IFN-alpha and IFN-beta), and neither type II IFN (IFN-gamma) nor various other cytokines enhanced TRAIL expression on anti-CD3-stimulated PBT cells. Type I IFNs have been used for clinical treatment of renal cell carcinomas (RCCs), and we found that most RCC cell lines were susceptible to TRAIL-induced apoptosis. Type I IFNs substantially augmented cytotoxic activity of anti-CD3-stimulated PBT cells against RCC cell lines in a TRAIL-dependent manner. These results indicate a unique feature of type I IFNs to regulate TRAIL-mediated T cell cytotoxicity, which may be involved in the antitumor effects of type I IFNs against various tumors.


Subject(s)
Antineoplastic Agents/metabolism , Apoptosis , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Interferon-alpha/metabolism , Interferon-beta/metabolism , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins , CD3 Complex/metabolism , Cytokines/metabolism , Cytokines/pharmacology , Cytotoxicity, Immunologic , Gene Expression Regulation/drug effects , Humans , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Jurkat Cells , Ligands , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , TNF-Related Apoptosis-Inducing Ligand , Transcription, Genetic/drug effects
11.
J Exp Med ; 172(6): 1877-80, 1990 Dec 01.
Article in English | MEDLINE | ID: mdl-2124255

ABSTRACT

The cytotoxic activity and pore-forming protein (PFP) expression of human peripheral blood (PB) gamma/delta T cells were examined. Fresh gamma/delta T cells isolated from PB lymphocytes by fluorescence-activated cell sorting exhibited a substantial natural killer-like cytotoxic activity against K562 target cells and had a high cytotoxic potential triggered by anti-CD3 monoclonal antibody (mAb) against P815 target cells bearing Fc gamma R. Immunocytochemical staining with an anti-PFP mAb revealed that virtually all PB gamma/delta T cells are granular lymphocytes with abundant PFP in their cytoplasmic granules. Constitutive expression of PFP in PB gamma/delta T cells was also demonstrated by Northern blot analysis. These observations support the proposed role of gamma/delta T cells in cytolytic immune surveillance in vivo.


Subject(s)
Cytotoxicity, Immunologic , Membrane Glycoproteins , Membrane Proteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Antibodies, Monoclonal , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD8 Antigens , Cells, Cultured , Humans , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Membrane Proteins/analysis , Perforin , Pore Forming Cytotoxic Proteins
12.
J Exp Med ; 170(4): 1445-50, 1989 Oct 01.
Article in English | MEDLINE | ID: mdl-2571670

ABSTRACT

By using mAb and flow cytometry, a constitutive expression of the p75 IL-2R was revealed in human peripheral blood CD8+ T cells and TCR delta-1+ T cells as well as in CD16+ NK cells. Anti-p75 IL-2R mAb almost completely inhibited the induction of cytolytic activity in these T cells by brief exposure to IL-2, as estimated by anti-TCR/CD3 mAb-targeted cytotoxicity. While anti-p55 IL-2R mAb alone inhibited the response only modestly, maximal inhibition was achieved by combining both anti-p55 and anti-p75 IL-2R mAbs. These results indicate that the p75 IL-2R constitutively expressed on peripheral blood CD8+ T cells and TCR delta-1+ T cells is predominantly responsible for the direct activation of these cells by IL-2.


Subject(s)
Lymphocyte Activation , Receptors, Interleukin-2/physiology , T-Lymphocytes/physiology , Antibodies, Monoclonal , Antigen-Antibody Reactions , CD4-Positive T-Lymphocytes/physiology , Cytotoxicity, Immunologic , Humans , In Vitro Techniques , Interleukin-2/pharmacology , Molecular Weight , Receptors, Interleukin-2/ultrastructure , T-Lymphocytes/classification
13.
J Exp Med ; 177(3): 863-8, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-7679713

ABSTRACT

Inflammatory cytokines have been implicated in the pathogenesis of rheumatoid arthritis (RA), whereas the mechanisms for constitutive production of inflammatory cytokines in affected joints are largely unknown. Recently, integrin-mediated interaction with extracellular matrix (ECM) proteins has been demonstrated to play a role in regulating cytokine production in T cells and monocytes. In this study, we investigated the contribution of the beta 1 integrin-mediated interaction with ECM proteins to the persistent cytokine gene expression in RA synovial fluid mononuclear cells (SFMNC). We examined mRNA expression of 14 cytokines in the SFMNC of three RA patients, which were either fresh or cultured overnight in serum-free medium on ECM-coated plates, by polymerase chain reaction with a panel of oligonucleotide primers specific for each cytokine. The persistent expression of various cytokine mRNA found in fresh SFMNC was maintained after overnight culture in serum-free medium on ECM proteins, especially on laminin (LM), but not on serum albumin. This effect of LM was inhibited by an anti-integrin beta 1 chain (CD29) mAb, as well as by an anti-CD3 mAb, indicating an important role of the beta 1 integrin-mediated interaction with ECM proteins in regulating persistent cytokine gene expression in RA SFMNC, and a key role of T cells in regulating inflammatory monokine production.


Subject(s)
Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cytokines/genetics , Extracellular Matrix Proteins/physiology , Gene Expression Regulation/genetics , Integrins/physiology , Synovial Fluid/cytology , Synovial Fluid/metabolism , Adult , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antigens, CD/pharmacology , Arthritis, Rheumatoid/genetics , CD3 Complex/immunology , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Cytokines/metabolism , Drug Interactions , Female , Gene Expression Regulation/drug effects , Humans , Integrin beta1 , Integrins/immunology , Laminin/pharmacology , Middle Aged , Monocytes/chemistry , Monocytes/cytology , Monocytes/metabolism , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/genetics , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
14.
J Exp Med ; 182(6): 1777-83, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7500022

ABSTRACT

Fas ligand (FasL) is a type II integral membrane protein homologous with tumor necrosis factor (TNF). Recent studies indicate that TNF is processed to yield the soluble cytokine by metalloproteinases at the cell surface of activated macrophages and T cells. In the present study, we investigated whether FasL is also released by metalloproteinases. Treatment with hydroxamic acid inhibitors of matrix metalloproteinases specifically led to accumulation of membrane-type FasL (p40) on the surface of human FasL cDNA transfectants and activated human T cells, as estimated by surface immunofluorescence and immunoprecipitation with newly established anti-human FasL monoclonal antibodies. This surface accumulation of mFasL was associated with the decrease of soluble FasL (p27) in the supernatant as estimated by quantitative ELISA and immunoprecipitation with anti-human FasL monoclonal antibodies. These results indicate that human FasL is efficiently released from the cell surface by metalloproteinases like TNF.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Fas Ligand Protein , Humans , Mice , Mice, Mutant Strains , Protease Inhibitors/pharmacology , Protein Processing, Post-Translational , Solubility , Transfection , Tumor Necrosis Factor-alpha/metabolism
15.
J Exp Med ; 176(5): 1241-9, 1992 Nov 01.
Article in English | MEDLINE | ID: mdl-1383383

ABSTRACT

CD2 is an intercellular adhesion molecule that has been implicated in T cell activation and differentiation both in humans and mice. Although the ligand for human CD2 has been defined as LFA-3, that for murine CD2 has not been identified yet. To identify the ligand for mouse CD2, we generated a chimeric molecule consisting of the extracellular domain of mouse CD2 and human immunoglobulin (Ig)G1 Fc (mCD2Rg). A hamster monoclonal antibody (mAb), HM48-1, was established by screening mAbs that could block the binding of mCD2Rg to T cell lines at the ligand site. The putative mouse CD2 ligand recognized by this mAb was a glycosyl phosphatidylinositol-anchored glycoprotein with an apparent molecular mass of 45 kD, which were shared characteristics with human LFA-3. However, its expression was predominantly restricted to hematopoietic cells, unlike human LFA-3. Protein microsequencing analysis for the NH2-terminal 18 amino acid residues of the affinity-purified HM48-1 antigen revealed that it is almost identical with mouse CD48. This identity was further confirmed by the reactivity of HM48-1 with a soluble recombinant CD48 (sCD48) protein and the molecule recognized by a rat mAb raised against sCD48. A rat anti-CD48 mAb blocked the mCD2Rg binding as well as HM48-1. Moreover, sCD48 also inhibited the mCD2Rg binding to the cellular ligand. Finally, like anti-CD2 mAb, HM48-1 inhibited the phytohemagglutinin response and, when crosslinked, augmented the anti-CD3 response of splenic T cells. These results indicate that CD48 is a ligand for mouse CD2 and is involved in regulating T cell activation.


Subject(s)
Antigens, CD/physiology , Antigens, Differentiation, T-Lymphocyte/metabolism , Lymphocyte Activation , Receptors, Immunologic/metabolism , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Base Sequence , CD2 Antigens , CD48 Antigen , CD58 Antigens , Cricetinae , Ligands , Male , Membrane Glycoproteins/physiology , Mice , Molecular Sequence Data , Rats , Recombinant Fusion Proteins/metabolism
16.
J Exp Med ; 193(6): 661-70, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11257133

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is expressed by in vitro activated natural killer (NK) cells, but the relevance of this observation to the biological function of NK cells has been unclear. Herein, we have demonstrated the in vivo induction of mouse TRAIL expression on various tissue NK cells and correlated NK cell activation with TRAIL-mediated antimetastatic function in vivo. Expression of TRAIL was only constitutive on a subset of liver NK cells, and innate NK cell control of Renca carcinoma hepatic metastases in the liver was partially TRAIL dependent. Administration of therapeutic doses of interleukin (IL)-12, a powerful inducer of interferon (IFN)-gamma production by NK cells and NKT cells, upregulated TRAIL expression on liver, spleen, and lung NK cells, and IL-12 suppressed metastases in both liver and lung in a TRAIL-dependent fashion. By contrast, alpha-galactosylceramide (alpha-GalCer), a powerful inducer of NKT cell IFN-gamma and IL-4 secretion, suppressed both liver and lung metastases but only stimulated NK cell TRAIL-mediated function in the liver. TRAIL expression was not detected on NK cells from IFN-gamma-deficient mice and TRAIL-mediated antimetastatic effects of IL-12 and alpha-GalCer were strictly IFN-gamma dependent. These results indicated that TRAIL induction on NK cells plays a critical role in IFN-gamma-mediated antimetastatic effects of IL-12 and alpha-GalCer.


Subject(s)
Carcinoma, Renal Cell/secondary , Interferon-gamma/immunology , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Liver Neoplasms/secondary , Membrane Glycoproteins/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Apoptosis Regulatory Proteins , Carcinoma, Renal Cell/immunology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Galactosylceramides/immunology , Galactosylceramides/pharmacology , Humans , Interleukin-12/immunology , Interleukin-12/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Kidney Neoplasms/pathology , Kidney Neoplasms/prevention & control , Ligands , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , TNF-Related Apoptosis-Inducing Ligand , Tissue Distribution , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis
17.
J Exp Med ; 183(4): 1473-81, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8666905

ABSTRACT

We assessed the role of CD40-CD40L, cytotoxic T lymphocyte (CTL)A4/CD28-B7s, and CD2-CD48/CD58 lymphocyte costimulatory pathways in the development of mercury chloride (HgCl2)-induced autoimmune disease in mice, which is believed to be mediated by T helper (Th) subset Th2. Inhibition of CD40-CD40-L and CTLA4/CD28-B7s interactions by anti-CD40-L antibody and soluble CTLA4-immunoglobulin (Ig) fusion protein, respectively, abrogated the autoimmune disease without affecting interleukin 4 (IL-4) production, showing the importance of physical contact between T and B lymphocytes in the Th2-mediated process. In contrast, two anti-CD2 antibodies that have been shown to induce immunosuppression of Th1-mediated events exacerbated the autoantibody response and augmented IgG1, IgE, and IL-4 production, transforming a mild mesangial glomerulopathy into a severe systemic immune complex disease. These observations demonstrate that manipulation of lymphocyte accessory counterreceptor interactions may affect the course of Th2-associated autoimmune disease and suggest that signals resulting from CD2 engagement play an essential role in the regulation of the Th1-Th2 effector equilibrium.


Subject(s)
Autoimmune Diseases/immunology , Cytotoxicity, Immunologic , Signal Transduction , Th2 Cells/immunology , Animals , Autoimmune Diseases/chemically induced , Base Sequence , Blood Vessels/pathology , CD2 Antigens/metabolism , CD40 Antigens/metabolism , CD40 Ligand , Cytokines/biosynthesis , Immune Complex Diseases , Membrane Glycoproteins/metabolism , Mercuric Chloride/pharmacology , Mice , Mice, Inbred Strains , Molecular Sequence Data , Skin/metabolism , Spleen/metabolism , T-Lymphocyte Subsets/immunology
18.
J Exp Med ; 184(1): 113-20, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8691124

ABSTRACT

Signals through the B cell antigen receptor lead to a variety of cellular events such as activation, anergy, and apoptosis. B cells select these outcomes to establish and maintain self-tolerance, and to mount adequate antibody responses. However, it is not fully understood how one and the same signal causes such different consequences. In the present study, we have studied the effect of activation signals on the outcome of responses to antigen receptor ligation. Two distinct growth-promoting signals were used to activate B cells. Ligation of either RP105, a newly discovered B cell surface molecule, or the CD40 molecule, drove B cells to proliferate. Resultant blastic cells were then exposed to anti-immunoglobulin M (IgM). Blast cells that had been stimulated with anti-RP105 ceased growing and underwent apoptosis after cross-linking of surface IgM. Coligation of the Fc gamma receptor IIB with surface IgM augmented, rather than aborted, this response. In contrast to RP105-activated B cells, blast cells that had been activated by CD40 ligation were unaltered by anti-IgM. On the other hand, CD40-activated B cells became extremely susceptible to Fas-mediated apoptosis, whereas RP105-activated B cells were much less sensitive. Anti-IgM-induced apoptosis in RP105 blasts was independent of Fas, because it was demonstrable with Fas-deficient MRL-lpr/lpr mice. These results demonstrate that the nature of an initial activation signal has a great influence on the fate of activated B cells after (re)engagement of the antigen receptor. RP105, as well as CD40, may be important in this life/death decision.


Subject(s)
Antigens, CD , Apoptosis , B-Lymphocytes/cytology , CD40 Antigens/physiology , Lymphocyte Activation , Membrane Proteins/physiology , Receptors, Antigen, B-Cell/physiology , Animals , Cell Cycle , Immunoglobulin M/physiology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Receptor Aggregation , Signal Transduction , fas Receptor/physiology
19.
J Exp Med ; 171(4): 1269-81, 1990 Apr 01.
Article in English | MEDLINE | ID: mdl-1691263

ABSTRACT

Our studies have analyzed pore-forming protein (PFP) mRNA expression in resting and stimulated human peripheral blood CD3- large granular lymphocytes (LGL), CD3+ T cells, and their CD4+ or CD8+ subsets. Signals that stimulate T cells to develop cytotoxic activity (i.e., IL-2 or OKT-3 mAb) led to the induction of PFP mRNA in T cells. The data indicated that IL-2 directly increased PFP mRNA in the CD8+ subset of T cells, in the absence of new DNA or protein synthesis. Abrogation of IL-2-induced PFP mRNA expression and cytotoxic potential of T cells by the anti-p75 IL-2 receptor mAb suggested that low numbers of p75 IL-2 receptors on CD8+ T cells were capable of transducing signals responsible for these IL-2-induced effects. The induction of T cell PFP mRNA via CD3, using OKT-3 mAb, was less rapid but greater than that caused by IL-2; however, a combination of PMA and ionomycin, which bypasses crosslinking of the TCR/CD3 complex, could not mimic this increase in PFP mRNA levels in T cells. The role of second messenger systems in regulating PFP mRNA expression remains to be determined. In contrast, high constitutive PFP mRNA expression was observed in CD3- LGL and these mRNA levels could not be enhanced by stimulation with IL-2. The cytotoxic potential of peripheral blood T cells and LGL induced in response to IL-2 correlated with IL-2-induced PFP mRNA levels in these cells and was consistent with PFP being one of several important molecules involved in the effector function of cytotoxic lymphocytes.


Subject(s)
Gene Expression/drug effects , Interleukin-2/pharmacology , Membrane Glycoproteins , Membrane Proteins/genetics , T-Lymphocytes, Cytotoxic/metabolism , Antibodies, Monoclonal , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD3 Complex , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Interferon-gamma/biosynthesis , Kinetics , Lymphocyte Activation , Perforin , Pore Forming Cytotoxic Proteins , RNA/blood , RNA/genetics , RNA/isolation & purification , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
20.
J Exp Med ; 191(2): 375-80, 2000 Jan 17.
Article in English | MEDLINE | ID: mdl-10637281

ABSTRACT

Infection of inbred mouse strains with Leishmania major is a well characterized model for analysis of T helper (Th)1 and Th2 cell development in vivo. In this study, to address the role of costimulatory molecules CD27, CD30, 4-1BB, and OX40, which belong to the tumor necrosis factor receptor superfamily, in the development of Th1 and Th2 cells in vivo, we administered monoclonal antibody (mAb) against their ligands, CD70, CD30 ligand (L), 4-1BBL, and OX40L, to mice infected with L. major. Whereas anti-CD70, anti-CD30L, and anti-4-1BBL mAb exhibited no effect in either susceptible BALB/c or resistant C57BL/6 mice, the administration of anti-OX40L mAb abrogated progressive disease in BALB/c mice. Flow cytometric analysis indicated that OX40 was expressed on CD4(+) T cells and OX40L was expressed on CD11c(+) dendritic cells in the popliteal lymph nodes of L. major-infected BALB/c mice. In vitro stimulation of these CD4(+) T cells showed that anti-OX40L mAb treatment resulted in substantially reduced production of Th2 cytokines. Moreover, this change in cytokine levels was associated with reduced levels of anti-L. major immunoglobulin (Ig)G1 and serum IgE. These results indicate that anti-OX40L mAb abrogated progressive leishmaniasis in BALB/c mice by suppressing the development of Th2 responses, substantiating a critical role of OX40-OX40L interaction in Th2 development in vivo.


Subject(s)
Antigens, CD , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Receptors, Tumor Necrosis Factor/immunology , Th1 Cells/immunology , Th2 Cells/immunology , 4-1BB Ligand , Animals , Antibodies, Monoclonal/immunology , CD27 Ligand , CD30 Ligand , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Disease Models, Animal , Female , Immunoglobulin G/immunology , Immunophenotyping , Ki-1 Antigen/immunology , Ligands , Lymphocytes/immunology , Membrane Glycoproteins/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , OX40 Ligand , Receptors, Immunologic , Receptors, OX40 , Th1 Cells/cytology , Th2 Cells/cytology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factors
SELECTION OF CITATIONS
SEARCH DETAIL