Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Clin Exp Rheumatol ; 20(6): 791-8, 2002.
Article in English | MEDLINE | ID: mdl-12508770

ABSTRACT

BACKGROUND: Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) induces apoptosis of tumor cells but not normal cells; its role in normal non-transformed tissues is unknown. OBJECTIVE: To evaluate the role of apoptosis mediated by TRAIL and TRAIL-receptor (TRAIL-R) system in lymphocytic sialadenitis in patients with Sjögren's syndrome. METHODS: The expression of TRAIL and TRAIL-R1, 2, 3 and 4 in lymphocytic sialadenitis was examined by immunoperoxidase staining in patients with Sjögren's syndrome and in normal subjects. To elucidate the mechanism of de novo expression of TRAIL-R1 antigen, we quantitatively investigated its induction by cytokines in human salivary duct cell line (HSG) by cell enzyme-linked immunosorbent assay. In human salivary duct cells stimulated by cytokines, we investigated the induction of apoptotic cell death by recombinant TRAIL protein. RESULTS: In patients with massive mononuclear cell infiltration, some infiltrating cells showed TRAIL. In patients with severe lymphocytic sialadenitis, TRAIL-R1, TRAIL-R2, or both were strongly expressed on the ductal epithelial cells. Neither TRAIL-R3 nor R4 were observed on ductal epithelium. In contrast, TRAIL-R1 and R2 were not found in the minor salivary glands of normal subjects or patients with mild lymphocytic sialadenitis. Unstimulated HSG cells did not express TRAIL-R1. Interferon-gamma (IFN-gamma) consistently upregulated levels of TRAIL-R1. In contrast, tumor necrosis factor-alpha (TNF-alpha), interleukin 1-beta (IL-1 beta), IL-2, and IL-4 had no effect on TRAIL-R1 levels. HSG cells expressing TRAIL-R1 in response to IFN-gamma were susceptible to apoptosis by recombinant TRAIL protein. CONCLUSION: Our findings suggest that TRAIL and TRAIL-R system may play a role in the pathogenesis of lymphocytic sialadenitis in patients with Sjögren's syndrome.


Subject(s)
Apoptosis , Membrane Glycoproteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Salivary Glands, Minor/metabolism , Sialadenitis/metabolism , Sjogren's Syndrome/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique, Indirect , Humans , Immunoenzyme Techniques , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Membrane Glycoproteins/pharmacology , Middle Aged , Recombinant Proteins/pharmacology , Salivary Glands, Minor/pathology , Sialadenitis/etiology , Sialadenitis/pathology , Sjogren's Syndrome/complications , Sjogren's Syndrome/pathology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/pharmacology
2.
Eur J Immunol ; 31(11): 3138-46, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11745330

ABSTRACT

Natural killer (NK) cells play an important role in early defense against viral infection. The cytotoxic activity of NK cells is increased by interferon-alpha/beta (IFN-alpha/beta), produced en masse in virally infected cells. However, the mechanism(s) by which IFN-alpha/beta contribute to the NK-cell-mediated antiviral response is not well understood. Here we provide evidence that the cytotoxicity of NK cells is enhanced by IFN-alpha/beta through induction of TNF-related apoptosis-inducing ligand (TRAIL). Isolation and analysis of the murine TRAIL promoter revealed the presence of an IFN-stimulated response element (ISRE), which binds to the transcription factor ISGF3 (interferon stimulated gene factor-3). This promoter is indeed activated by IFN-beta in ISGF3-dependent manner. We also show that virally infected cells, but not uninfected cells, are susceptible to TRAIL-mediated cytotoxicity in vitro, and that the TRAIL expressed in NK cells is indeed crucial in limiting virus replication in vivo. Thus, our study reveals a new molecular link between IFN-alpha/beta signaling and activation of NK cells in antiviral response of the host.


Subject(s)
Cardiovirus Infections/immunology , Cytotoxicity, Immunologic , Encephalomyocarditis virus/immunology , Interferon-alpha/physiology , Interferon-beta/physiology , Killer Cells, Natural/immunology , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Apoptosis Regulatory Proteins , DNA-Binding Proteins/physiology , Gene Expression Regulation , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, SCID , TNF-Related Apoptosis-Inducing Ligand , Transcription Factors/physiology , Transcriptional Activation , Tumor Necrosis Factor-alpha/physiology
3.
Ann Surg ; 234(5): 681-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11685033

ABSTRACT

OBJECTIVE: To investigate the role of FADD (Fas-associated protein with death domain) in Fas and tumor necrosis factor receptor 1 (TNFR1)-mediated hepatic injury and inflammatory response in vivo. SUMMARY BACKGROUND DATA: Fas and TNFR1 are cell surface molecules that trigger apoptosis or inflammation on engagement by a specific ligand or antibody. FADD is recruited to the cytoplasmic domain of these receptors on their activation and works as a common mediator to induce apoptosis. It is known that a blockade of FADD can inhibit apoptosis mediated by Fas or TNFR1 in vitro. However, it is not known whether the blockade can prevent organ injury and whether the inflammatory cascade is affected in vivo. METHODS: A FADD deletion mutant lacking the death effector domain was introduced into mice by transduction with an adenovirus vector, and the effect of this FADD dominant negative mutant was examined in several liver injury models. RESULTS: Hepatic injury induced by anti-Fas monoclonal antibody or tumor necrosis factor (TNF)-alpha plus D-galactosamine was markedly ameliorated by the FADD dominant negative transduction, which abrogated the death rate. Further, the FADD dominant negative transduction efficiently blocked T cell- mediated concanavalin A-induced hepatitis while not affecting TNF-alpha production or TNF-alpha-induced nuclear factor-kappaB activation in the liver. CONCLUSIONS: These results provide the basis for a novel therapeutic modality in which an unfavorable apoptotic process can be inhibited without affecting a favorable response for liver regeneration; this would be relevant to the clinical treatment of acute and chronic liver diseases as well as to some inflammatory disorders with hypercytokinemia, such as sepsis.


Subject(s)
Adaptor Proteins, Signal Transducing , Antigens, CD/physiology , Apoptosis/physiology , Carrier Proteins/physiology , Membrane Glycoproteins/physiology , NF-kappa B/physiology , Receptors, Tumor Necrosis Factor/physiology , Adenoviridae , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD/immunology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Concanavalin A , Fas Ligand Protein , Fas-Associated Death Domain Protein , Galactosamine/pharmacology , Gene Transfer Techniques , Genetic Vectors , Hepatitis/etiology , Hepatitis/physiopathology , Liver/metabolism , Liver/pathology , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha/analysis
4.
J Immunol ; 167(6): 3266-75, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11544314

ABSTRACT

Fas ligand (L)/CD95L, a proapoptotic member of the TNF family, is a potential target for clinical intervention in various diseases. In the present study, we generated a humanized anti-human FasL mAb and characterized the epitopes of neutralizing mAbs by extensive alanine-scanning mutagenesis of human FasL. The predicted molecular model of FasL trimer revealed that the mAbs recognize largely overlapped conformational epitopes that are composed of two clusters, one around the outer tip-forming D-E loop and another near the top of FasL. Both of these sites on FasL are critically involved in the direct interaction with the corresponding receptor, Fas. These results suggest that the mAbs efficiently neutralize FasL cytotoxicity by masking both of these FasL/Fas contact sites.


Subject(s)
Antibodies, Monoclonal/immunology , Epitope Mapping , Membrane Glycoproteins/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibody Specificity , Apoptosis , Binding Sites, Antibody , CHO Cells , COS Cells , Chlorocebus aethiops , Computer Simulation , Cricetinae , Cricetulus , Cytotoxicity, Immunologic , Epitopes/chemistry , Epitopes/immunology , Fas Ligand Protein , Gene Rearrangement, B-Lymphocyte , Genes, Immunoglobulin , Humans , Macromolecular Substances , Membrane Glycoproteins/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Neutralization Tests , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Structure-Activity Relationship , fas Receptor/immunology
5.
Cytokine ; 14(4): 193-201, 2001 May 21.
Article in English | MEDLINE | ID: mdl-11448118

ABSTRACT

Interferon-alpha (IFN-alpha) exerts the anti-tumour effect on various tumours at least partly through induction of apoptosis. Apoptosis is induced by members of the tumour necrosis factor (TNF) family, including Fas (CD95) and TNF-related apoptosis-inducing ligand (TRAIL). In the present study, we examined whether the TRAIL/TRAIL-R system is involved in IFN-alpha-induced apoptosis using Daudi B lymphoma cells. IFN-alpha upregulated the expression of TRAIL within 12 h, as assessed by flow cytometry and RT-PCR, and the level increased with time until 72 h. The levels of both TRAIL-R1 and TRAIL-R2, low in Daudi cells, were enhanced by IFN-alpha. The enhanced TRAIL-R1/-R2 appeared to function as a death-inducing molecule since IFN-alpha-stimulated cells were more susceptible to TRAIL-induced cell death. The IFN-alpha-stimulated Daudi cells or their derived culture supernatants displayed cytotoxicity against TRAIL-sensitive, but not resistant lines. Moreover, the IFN-alpha-induced reduction in mitochondrial membrane potential preceding the induction of apoptosis was substantially prevented by neutralizing anti-TRAIL monoclonal antibody. Taken together, IFN-alpha-induced apoptosis appears to be mediated by the autocrine and/or paracrine loop involving TRAIL/TRAIL-R.


Subject(s)
Apoptosis/immunology , Interferon-alpha/pharmacology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Membrane Glycoproteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antibodies, Monoclonal/pharmacology , Apoptosis Regulatory Proteins , Cell Death/immunology , Growth Inhibitors/pharmacology , Humans , Immunity, Innate , Interferon-alpha/antagonists & inhibitors , Lymphoma, B-Cell/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , RNA, Messenger/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/immunology
6.
Biochem Biophys Res Commun ; 284(5): 1162-7, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11414705

ABSTRACT

To elucidate the molecular mechanism(s) involved in the TRAIL-induced apoptosis sensitivity, we conducted the following experiments utilizing TRAIL-sensitive and -resistant glioma cells. We examined the expression of TRAIL receptors mRNA, but no significant differences were detected in those cells. TRAIL-resistant cells were sensitized to TRAIL-induced apoptosis by staurosporine pretreatment and preferentially expressed PKCepsilon. Since several lines of evidence suggest that PKC may play a protective role for apoptosis, we analyzed the involvement of PKCepsilon in TRAIL-induced apoptosis by an adenovirus vector expression system. We found that TRAIL susceptibility was augmented by the expression of a dominant negative PKCepsilon in TRAIL-resistant cells. Conversely, PKCepsilon introduction in TRAIL-sensitive cells resulted in the reduction of TRAIL-induced apoptosis. Taken together, these data suggest that PKCepsilon may be a regulator of susceptibility to TRAIL-induced apoptosis in gliomas and probably other malignancies.


Subject(s)
Apoptosis , Isoenzymes/pharmacology , Membrane Glycoproteins/pharmacology , Protective Agents/pharmacology , Protein Kinase C/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Cell Survival/drug effects , Drug Interactions , Glioma , Humans , Protein Kinase C-epsilon , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured
7.
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
8.
J Exp Med ; 193(5): 651-60, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11238596

ABSTRACT

Apoptosis is a key for CD4+ T cell destruction in HIV-1-infected patients. In this study, human peripheral blood lymphocyte (PBL)-transplanted nonobese diabetic (NOD)-severe combined immunodeficient (SCID) (hu-PBL-NOD-SCID) mice were used to examine in vivo apoptosis after HIV-1 infection. As the hu-PBL-NOD-SCID mouse model allowed us to see extensive infection with HIV-1 and to analyze apoptosis in human cells in combination with immunohistological methods, we were able to quantify the number of apoptotic cells with HIV-1 infection. As demonstrated by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), massive apoptosis was predominantly observed in virus-uninfected CD4+ T cells in the spleens of HIV-1-infected mice. A combination of TUNEL and immunostaining for death-inducing tumor necrosis factor (TNF) family molecules indicated that the apoptotic cells were frequently found in conjugation with TNF-related apoptosis-inducing ligand (TRAIL)-expressing CD3+CD4+ human T cells. Administration of a neutralizing anti-TRAIL mAb in HIV-1-infected mice markedly inhibited the development of CD4+ T cell apoptosis. These results suggest that a large number of HIV-1-uninfected CD4+ T cells undergo TRAIL-mediated apoptosis in HIV-infected lymphoid organs.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/metabolism , HIV Infections/immunology , HIV-1/immunology , Membrane Glycoproteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , CD3 Complex/biosynthesis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Crosses, Genetic , Disease Models, Animal , Graft Survival , HIV-1/pathogenicity , Humans , In Situ Nick-End Labeling , Lymphocyte Transfusion , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Spleen/metabolism , Spleen/pathology , Spleen/virology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology
9.
Blood ; 97(7): 2067-74, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264173

ABSTRACT

Human Valpha24NKT cells are activated by alpha-galactosylceramide (alpha-GalCer)-pulsed dendritic cells in a CD1d-dependent and a T-cell receptor-mediated manner. Here, we demonstrate that CD4(+)V alpha 24NKT cells derived from a patient with acute myeloid leukemia (AML) M4 are phenotypically similar to those of healthy donors and, in common with those derived from healthy donors, express tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) when the cells are activated by alpha-GalCer-pulsed dendritic cells but not prior to activation. We also show that myeloid leukemia cells from patients with AML M4, but not from patients with AML M0 or M1, undergo apoptosis following culture with TRAIL-expressing autologous or allogeneic healthy donor V alpha 24NKT cells. Apoptosis of AML M4 leukemia cells from patient peripheral blood was almost completely blocked by a neutralizing monoclonal antibody against TRAIL, indicating that TRAIL on V alpha 24NKT cells is essential for the induction of apoptosis in AML M4 leukemia cells. A nonobese diabetic-severe combined immunodeficient human leukemia (AML M4) model showed that human activated CD4(+)V alpha 24NKT cells induced apoptosis of human leukemia cells in vivo. This is the first evidence that activated V alpha 24NKT cells express TRAIL and that TRAIL causes apoptosis of monocytic leukemia cells from patients with AML M4 in vitro and in vivo. Adoptive immune therapy with activated V alpha 24NKT cells, or other strategies to increase activated V alpha 24NKT cells in vivo, may be of benefit to patients with AML M4. (Blood. 2001;97:2067-2074)


Subject(s)
Apoptosis/physiology , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Galactosylceramides/pharmacology , Immunotherapy, Adoptive , Leukemia, Myelomonocytic, Acute/pathology , Membrane Glycoproteins/physiology , T-Lymphocyte Subsets/metabolism , Tumor Necrosis Factor-alpha/physiology , Adult , Animals , Antibodies, Monoclonal/immunology , Antigens, CD1/physiology , Antigens, CD1d , Apoptosis Regulatory Proteins , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cells, Cultured , Coculture Techniques , Female , Fetal Blood/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid/pathology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/immunology , Leukemia, Myelomonocytic, Acute/therapy , Lymphocyte Activation , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Xenograft Model Antitumor Assays
10.
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
11.
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
12.
J Invest Dermatol ; 115(2): 154-61, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10951230

ABSTRACT

Fas/Fas ligand-mediated lymphocyte apoptosis has been implicated in the suppression of immune responses and may cause immune privilege. Human corneas exhibit immune privilege and can be transplanted across allogeneic barriers without immunosuppressive therapy, perhaps, because corneal keratinocytes express Fas ligand. To characterize Fas and Fas ligand expression in skin, we examined expression by murine keratinocytes, dermal fibroblasts, melanocytes, and human umbilical endothelial cells. We also studied the regulation of Fas and Fas ligand in skin cells by retinoic acid, vitamin D3, and dexamethasone as well as various cytokines. Among the molecules and cells tested, retinoic acid selectively upregulated the expression of Fas ligand molecule by fibroblasts. Retinoic acid-induced Fas ligand+ fibroblasts killed Fas+ target cells, and this killing was blocked by anti-Fas ligand antibody. The function of Fas ligand on dermal fibroblasts in vivo was tested in a cutaneous allograft system. Histoincompatible BALB/C mouse (H-2d) donor skin was grafted on to allogeneic C57BL/6 mice (H-2b). Daily local injection of retinoic acid blocked inflammation and extended graft survival for more than 10 d. Injection of retinoic acid into Fas ligand mutated gld/gld donor skin did not prevent leukocyte infiltration into the allograft or prolong graft survival. These experiments indicate that, in skin, retinoic acid selectively increases Fas ligand expression by fibroblasts and that retinoic acid has potent Fas/Fas ligand-dependent immunosuppressive activity.


Subject(s)
Fibroblasts/metabolism , Graft Survival/drug effects , Membrane Glycoproteins/physiology , Skin Transplantation/immunology , Tretinoin/pharmacology , fas Receptor/physiology , Animals , Cell Death/physiology , Cell Membrane/metabolism , Cells, Cultured , Fas Ligand Protein , Humans , Jurkat Cells/metabolism , Jurkat Cells/physiology , Keratinocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mutation/physiology , Reference Values , Skin/cytology , Skin/drug effects , Skin/immunology , Skin/metabolism , Tissue Donors , Up-Regulation , fas Receptor/genetics
13.
J Immunol ; 165(2): 997-1003, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10878376

ABSTRACT

To study the pathophysiological roles of overexpressed caspase-1 (CASP1), originally designated as IL-1 beta-converting enzyme, we generated transgenic mice in which human CASP1 is overexpressed in their keratinocytes. The transgenic mice spontaneously developed recalcitrant dermatitis and skin ulcers, characterized by the presence of massive keratinocyte apoptosis. The skin of the mice contained the active form of human CASP1 and expressed mRNA for caspase-activated DNase, an effector endonuclease responsible for DNA fragmentation. Their skin and sera showed elevated levels of mature IL-18 and IL-1 beta, but not of IFN-gamma. The plasma from these animals induced IFN-gamma production by IL-18-responsive NK cells. Administration of heat-killed Propionibacterium acnes, a potent in vivo type 1 cell inducer, caused IFN-gamma-mediated lethal liver injury in the transgenic mice, which was completely inhibited by treatment with neutralizing anti-IL-18 Ab. These results indicated that in vivo overexpression of CASP1 caused spontaneous apoptotic tissue injury and rendered mice highly susceptible to exogenous type 1 cell-inducing condition in collaboration with endogenously accumulated proinflammatory cytokines.


Subject(s)
Apoptosis/genetics , Caspase 1/genetics , Interleukin-18/blood , Shock, Septic/enzymology , Shock, Septic/pathology , Skin/enzymology , Skin/pathology , Animals , Apoptosis/immunology , Caspase 1/biosynthesis , Caspase 1/metabolism , Crosses, Genetic , Enzyme Activation , Female , Hepatitis, Animal/enzymology , Hepatitis, Animal/microbiology , Hepatitis, Animal/pathology , Humans , Keratinocytes/enzymology , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Propionibacterium acnes , Shock, Septic/genetics , Shock, Septic/immunology , Skin/immunology , Skin Ulcer/enzymology , Skin Ulcer/genetics , Skin Ulcer/pathology , Substrate Specificity/genetics
14.
J Autoimmun ; 14(2): 151-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10677246

ABSTRACT

Since Fas ligand (FasL) can induce apoptosis of Fas-bearing cells, Fas/FasL interactions can play a critical role in maintaining self-tolerance. Fas/FasL interactions in lupus-like autoimmune disease have been well characterized in studies using either Fas or FasL mutant mice. However, the effect of the defective FasL-mediated signaling on the establishment of lupus in other mouse strains, such as NZB/W (B/W) F1, remains uncertain. In the present study, we examined the effect of anti-FasL monoclonal antibody (mAb) on the development of lupus. Treatment of B/W F1 mice with anti-FasL mAb augmented IgG1- and IgG2a-type anti-dsDNA Ab production. However, treatment of B/W F1 mice with anti-FasL mAb also significantly prevented the development of lupus nephritis. These results indicate that Fas/FasL interactions not only regulate IgG-type autoantibody production, but also influence the development of lupus nephritis in B/W F1 mice.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lupus Nephritis/immunology , Lupus Nephritis/prevention & control , Membrane Glycoproteins/antagonists & inhibitors , Animals , Antibodies, Antinuclear/blood , Fas Ligand Protein , Female , Hybridization, Genetic , Immunoglobulin G/blood , Lupus Nephritis/blood , Lymphocyte Count , Membrane Glycoproteins/immunology , Mice , Mice, Inbred NZB
15.
J Immunol ; 164(6): 2931-6, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10706679

ABSTRACT

Previous reports that diabetogenic lymphocytes did not induce diabetes in nonobese diabetic (NOD)-lpr mice suggested the critical role of Fas-Fas ligand (FasL) interaction in pancreatic beta cell apoptosis. However, recent works demonstrated that FasL is not an effector molecule in islet beta cell death. We addressed why diabetes cannot be transferred to NOD-lpr mice despite the nonessential role of Fas in beta cell apoptosis. Lymphocytes from NOD-lpr mice were constitutively expressing FasL. A decrease in the number of FasL+ lymphocytes by neonatal thymectomy facilitated the development of insulitis. Cotransfer of FasL+ lymphocytes from NOD-lpr mice completely abrogated diabetes after adoptive transfer of lymphocytes from diabetic NOD mice. The inhibition of diabetes by cotransferred lymphocytes was reversed by anti-FasL Ab, indicating that FasL on abnormal lymphocytes from NOD-lpr mice was responsible for the inhibition of diabetes transfer. Pretreatment of lymphocytes with soluble FasL (sFasL) also inhibited diabetes transfer. sFasL treatment decreased the number of CD4+CD45RBlow cells and increased the number of propidium iodide-stained cells among CD4+CD45RBlow cells, suggesting that sFasL induces apoptosis on CD4+CD45RBlow "memory" cells. These results resolve the paradox between previous findings and suggest a new role for FasL in the treatment of autoimmune disorders. Our data also suggest that sFasL is involved in the deletion of potentially hazardous peripheral "memory" cells, contrary to previous reports that Fas on unmanipulated peripheral lymphocytes is nonfunctional.


Subject(s)
Apoptosis/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Membrane Glycoproteins/physiology , fas Receptor/metabolism , Adoptive Transfer , Animals , Diabetes Mellitus, Type 1/pathology , Fas Ligand Protein , Female , Immunologic Memory/immunology , Ligands , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Inbred NOD , Solubility , Spleen/cytology , Spleen/immunology
16.
J Dermatol Sci ; 22(2): 96-101, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10674822

ABSTRACT

Epidermal Langerhans cells (LC) are potent antigen-presenting cells (APC), that play a crucial role in initiating cutaneous immune responses. The Fas/Fas ligand pathway has been implicated as an important cellular pathway in the regulation of peripheral immunity. The morphologic, functional and phenotypic characteristics of LC are becoming well-characterized. However, the mechanisms involved in eliminating LC are poorly understood. In this report, we demonstrated that murine epidermal LC constitutively express the Fas antigen (CD95) and the expression was up-regulated by the addition of IFN-gamma in cultures. Interestingly, epidermal LC underwent apoptosis by the addition of both recombinant soluble Fas ligand (FasL) and IFN-gamma, but not by FasL alone. These results suggest that LC may acquire the susceptibility to Fas-mediated apoptosis through up-regulation of the Fas expression by IFN-gamma derived from activated T cells and that the elimination of LC may be important for preventing excess cutaneous inflammatory diseases.


Subject(s)
Apoptosis , Langerhans Cells/pathology , Membrane Glycoproteins , fas Receptor , Animals , Antigen Presentation , Apoptosis/immunology , Cells, Cultured , Fas Ligand Protein , Langerhans Cells/immunology , Membrane Glycoproteins/immunology , Mice , Signal Transduction/immunology , fas Receptor/immunology
17.
Immunity ; 11(3): 359-67, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10514014

ABSTRACT

IL-18, produced as a biologically inactive precursor, is processed by caspase-1 in LPS-activated macrophages. Here, we investigated caspase-1-independent processing of IL-18 in Fas ligand (FasL)-stimulated macrophages and its involvement in liver injury. Administration of Propionibacterium acnes (P. acnes) upregulated functional Fas expression on macrophages in an IFNgamma-dependent manner, and these macrophages became competent to secrete mature IL-18 upon stimulation with FasL. This was also the case for caspase-1-deficient mice. Administration of recombinant soluble FasL (rFasL) after P. acnes priming induced comparable elevation of serum IL-18 in parallel with elevated serum liver enzyme levels. However, liver injury was not induced in IL-18-deficient mice after rFasL administration. These results indicate a caspase-1-independent pathway of IL-18 secretion from FasL-stimulated macrophages and its critical involvement in FasL-induced liver injury.


Subject(s)
Caspase 1/metabolism , Interleukin-18/metabolism , Liver Diseases/immunology , Macrophages/metabolism , Membrane Glycoproteins/metabolism , fas Receptor/metabolism , Amino Acid Chloromethyl Ketones , Animals , Caspase 1/genetics , Caspase Inhibitors , Cells, Cultured , Cysteine Proteinase Inhibitors , Fas Ligand Protein , Female , Humans , Liver Diseases/pathology , Macrophages/drug effects , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Solubility
18.
Clin Exp Immunol ; 118(1): 108-14, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10540167

ABSTRACT

To determine the mechanisms responsible for the impaired lymphocyte-mediated cytotoxicity in Chediak-Higashi syndrome (CHS), we investigated the killing ability of peripheral blood lymphocytes (PBL) from three patients with CHS using several kinds of target cells that were sensitive to perforin, Fas ligand (FasL), and/or tumour necrosis factor-alpha (TNF-alpha). Freshly isolated CHS PBL did not kill K562 target cells, killing of which by normal PBL was perforin-dependent, as demonstrated by complete inhibition by concanamycin A (CMA), an inhibitor of perforin-based cytotoxicity. In contrast, the CHS PBL exhibited substantial cytotoxicity against Jurkat cells, which was only partially inhibited by CMA treatment but not by the addition of neutralizing anti-FasL or anti-TNF-alpha antibodies. IL-2-activated CHS PBL exhibited substantial levels of cytotoxicity against K562 and Jurkat cells, the levels being 74% and 83% of the respective normal control values, respectively. CMA treatment showed that while the cytotoxicity of IL-2-activated CHS PBL against K562 was largely dependent on perforin, that against Jurkat was largely not. IL-2-activated CHS PBL expressed FasL mRNA, and killed Fas transfectants. These findings indicate that CHS PBL have an ability to kill some target cells via a perforin-mediated pathway, especially when they are activated by IL-2. It was also demonstrated that CHS PBL can exert cytotoxicity against certain target cells by utilizing FasL and an undefined effector molecule other than perforin, FasL, or TNF-alpha.


Subject(s)
Chediak-Higashi Syndrome/immunology , Cytotoxicity, Immunologic/immunology , Macrolides , Adult , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis/immunology , Cells, Cultured , Chediak-Higashi Syndrome/pathology , Child , Child, Preschool , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic/drug effects , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Female , Flow Cytometry , Humans , Interleukin-2/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/pharmacology , RNA, Messenger/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
19.
Drug Des Discov ; 16(2): 119-30, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10533808

ABSTRACT

Both Fas ligand (FasL) and tumor necrosis factor-alpha (TNF-alpha) are type II integral membrane proteins. Recently, we have reported that FasL is processed to a soluble form by an unknown metalloproteinase at the cell surface and some hydroxamate matrix metalloproteinase (MMP) inhibitors inhibit the processing similar to the case observed with TNF-1alpha. We studied the inhibitory effects of various hydroxamate MMP inhibitors on FasL and TNF-alpha processing in order to characterize the processing enzymes using human FasL cDNA transfectants and LPS-stimulated THP-1 cells. It turned out that (1) the P1' group of hydroxamates was very important for the selective inhibitory activity toward TNF-alpha and FasL processing, (2) P1' 3-phenylpropyl group was favorable for the inhibition of FasL processing, and (3) P1' isobutyl and isopropyl groups were favorable for that of TNF-alpha processing. These differences in sensitivity to inhibitors imply that (1) membrane-bound FasL and TNF-alpha might be processed by distinct metalloproteinases, (2) the S1' site of FasL processing enzyme differs from that of MMP-1 and MMP-9, but appears to be similar to that of MMP-3, and (3) the S1' site of TNF-alpha processing enzyme is smaller than that of FasL processing enzyme. These results would be helpful in designing a more selective inhibitor.


Subject(s)
Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , fas Receptor/biosynthesis , Humans , Lipopolysaccharides/pharmacology , Structure-Activity Relationship , Transfection
20.
Br J Dermatol ; 141(2): 201-5, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10468788

ABSTRACT

Epidermal Langerhans cells (LC) are potent antigen-presenting cells (APC) that play a crucial part in initiating cutaneous immune responses. Although functional roles of LC as APC in the draining lymph node have been well investigated, little is known about the fate of LC after the antigen presentation to T cells. In this report, we demonstrate that antigen-bearing LC that migrated into the draining lymph nodes and were identified as fluorescent cells after skin painting with fluorescein isothiocyanate also expressed the Fas antigen. Clearance of the antigen-bearing LC was significantly delayed and the ratio of dead cells was reduced in Fas-deficient lpr and Fas ligand (FasL)-deficient gld mice at 2 days after skin painting, suggesting the involvement of a Fas/FasL-mediated pathway in the elimination of antigen-bearing LC in draining lymph nodes. These results suggest that a substantial population of LC after antigen presentation may undergo Fas/FasL-mediated apoptosis and that the elimination of active APC may be important for preventing excess immune responses.


Subject(s)
Langerhans Cells/immunology , Lymph Nodes/immunology , Membrane Glycoproteins/immunology , fas Receptor/immunology , Animals , Apoptosis/immunology , Fas Ligand Protein , Female , Fluorescein-5-isothiocyanate , Langerhans Cells/cytology , Langerhans Cells/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C3H
SELECTION OF CITATIONS
SEARCH DETAIL
...