Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38168302

ABSTRACT

Background: The inability to evaluate host immunity in a rapid quantitative manner in patients with sepsis has severely hampered development of novel immune therapies. The ELISpot assay is a functional bioassay that measures the number of cytokine-secreting cells and the relative amount of cytokine produced at the single-cell level. A key advantage of ELISpot is its excellent dynamic range enabling a more precise quantifiable assessment of host immunity. Herein, we tested the hypothesis on whether the ELISpot assay can detect dynamic changes in both innate and adaptive immunity as they often occur during sepsis. We also tested whether ELISpot could detect the effect of immune drug therapies to modulate innate and adaptive immunity. Methods: Mice were made septic using sublethal cecal ligation and puncture (CLP). Blood and spleens were harvested serially and ex vivo IFN-γ and TNF-α production were compared by ELISpot and ELISA. The capability of ELISpot to detect changes in innate and adaptive immunity due to in vivo immune therapy with dexamethasone, IL-7, and arginine was also evaluated. Results: ELISpot confirmed a decreased innate and adaptive immunity responsiveness during sepsis progression. More importantly, ELISpot was also able to detect changes in adaptive and innate immunity in response to immune-modulatory reagents, for example dexamethasone, arginine, and IL-7 in a readily quantifiable manner, as predicted by the reagents known mechanisms of action. ELISpot and ELISA results tended to parallel one another although some differences were noted. Conclusion: ELISpot offers a unique capability to assess the functional status of both adaptive and innate immunity over time. The results presented herein demonstrate that ELISpot can also be used to detect and follow the in vivo effects of drugs to ameliorate sepsis-induced immune dysfunction. This capability would be a major advance in guiding new immune therapies in sepsis.

2.
Cancer Gene Ther ; 14(12): 976-84, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17853923

ABSTRACT

High levels of decoy receptor 2 (DcR2; TRAIL-R4) expression are correlated with TRAIL resistance in prostate cancer cells. In addition, upregulation of TRAIL death receptor (DR4 and DR5) expression, either by ionizing radiation or chemotherapy, can sensitize cancer cells to TRAIL. Considering more than half of human cancers are TRAIL resistant, modulation of surface TRAIL receptor expression appears to be an attractive treatment modality to counteract TRAIL resistance. In this study, three siRNA duplexes targeting DcR2 receptor were tested. Ad5hTRAIL infections were performed to overexpress human full-length TRAIL to induce cell death, and the in vitro tumorigenic potential of prostate cancer cells was assessed using colony-forming assays on soft agar. The DU145 and LNCaP prostate cancer cell lines, which express high levels of DcR2, were resistant to Ad5hTRAIL-induced death. Downregulation of surface DcR2 expression by siRNA sensitized these prostate cancer cell lines to Ad5hTRAIL. In addition, DcR2 siRNA-mediated knockdown of DcR2, followed by Ad5hTRAIL infection, dramatically reduced the in vitro tumorigenic potential of prostate cancer cells. Collectively, our results suggest the potential for combining receptor-specific siRNA with TRAIL in the treatment of certain cancers.


Subject(s)
Adenoviridae , Genetic Therapy , Neoplasm Proteins/antagonists & inhibitors , Prostatic Neoplasms/metabolism , RNA, Small Interfering/biosynthesis , Transduction, Genetic , Tumor Necrosis Factor Decoy Receptors/antagonists & inhibitors , Animals , COS Cells , Cell Death/genetics , Cell Line, Tumor , Chlorocebus aethiops , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , RNA, Small Interfering/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics , Up-Regulation/genetics
3.
Cancer Gene Ther ; 13(6): 628-32, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16456549

ABSTRACT

Renal cell carcinoma (RCC) will cause greater than 12,000 deaths in the United States this year. The lack of effective therapy for disseminated RCC has stimulated the search for novel treatments including immunotherapeutic strategies, but poor therapeutic responses and marked toxicity have limited their use. The tumor necrosis factor (TNF) family member TNF-related apoptosis-inducing ligand (TRAIL)/Apo-2L induces apoptosis in various tumor cell types, while having little cytotoxicity against normal cells. In this study, we investigated the tumoricidal potential of a recombinant adenovirus encoding human TNFSF10 (Ad5-TRAIL), alone and in combination with a panel of histone deacetylase inhibitors (HDACi), against the TRAIL/Apo-2L-resistant RCC line 786-O and normal human renal proximal tubule epithelial cells (RPTEC). Ad5-TRAIL was unable to induce apoptosis in either 786-O or RPTEC alone; however, tumor cell apoptosis occurred when Ad5-TRAIL was combined with HDAC inhibition. Except when combined with trichostatin A, RPTEC were not sensitized to Ad5-TRAIL by HDACi. In 786-O, HDAC inhibition induced CAR expression, permitting increased adenoviral infection and transgene expression. It also induced TRAIL-R2 expression, accelerated the death-inducing signaling complex formation and enhanced caspase-8 activation. Our results demonstrate the utility of combining Ad5-TRAIL with HDACi against RCC, and mechanistically define how this combination modulates RCC sensitivity to TRAIL/Apo-2L and adenoviral infection.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/drug effects , Carcinoma, Renal Cell/drug therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Histone Deacetylase Inhibitors , Kidney Neoplasms/drug therapy , Membrane Glycoproteins/genetics , Tumor Necrosis Factor-alpha/genetics , Adenoviridae/classification , Adenoviridae/genetics , Antineoplastic Agents/administration & dosage , Apoptosis Regulatory Proteins/administration & dosage , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Caspase 8 , Caspases/metabolism , Cell Line, Tumor , Combined Modality Therapy , Enzyme Inhibitors/therapeutic use , Genetic Vectors/genetics , Histone Deacetylases/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Membrane Glycoproteins/administration & dosage , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/administration & dosage
4.
Apoptosis ; 8(6): 587-99, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14739604

ABSTRACT

A great deal of enthusiasm is being generated for TRAIL (TNF-related apoptosis-inducing ligand)/Apo-2L as a tumor therapeutic agent because it is cytotoxic to a variety of tumor cell types but not normal cells. Moreover, it is well documented that TRAIL/Apo-2L-induced tumor cell death is a caspase-dependent apoptotic process. Through the use of a transfected cell line expressing murine TRAIL/Apo-2L and a recombinant adenovirus encoding the murine TRAIL/Apo-2L cDNA (Ad5-mTRAIL) against two murine tumor cell lines [TRAMP-C2 (prostate adenocarcinoma) and Renca (renal adenocarcinoma)], we found that mTRAIL/Apo-2L also can kill tumor cells by inducing necrosis. Specifically, we observed the default method of mTRAIL/Apo-2L-induced death in TRAMP-C2 cells was via a necrotic process, characterized by the complete lack of an annexin V(+)/PI(-) population, SAPK/JNK phosphorylation, caspase activation, Bid cleavage, or cytochrome c release. Moreover, the inclusion of zVAD-fmk, an inhibitor of caspase activation, markedly enhanced mTRAIL/Apo-2L-mediated killing of TRAMP-C2. In contrast, apoptosis was induced in TRAMP-C2 using TNF, as measured by the criteria listed above, as was Renca by mTRAIL/Apo-2L. These results demonstrate the natural occurrence of both TRAIL/Apo-2L-induced apoptotic and necrotic signaling mechanisms within tumor cells.


Subject(s)
Apoptosis/physiology , Membrane Glycoproteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins , Membrane Glycoproteins/genetics , Mice , Microscopy, Electron , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
5.
Mol Ther ; 4(3): 257-66, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11545617

ABSTRACT

TRAIL is a member of the tumor necrosis factor superfamily that induces apoptosis in a variety of tumor cell types both in vitro and in vivo, while demonstrating minimal cytotoxicity toward normal tissues. One disadvantage to previous in vivo protocols was the need for large quantities of TRAIL to suppress tumor growth. Here we engineered a replication-deficient adenovirus to encode human TNFSF10 (Ad5-TRAIL) as an alternative to recombinant, soluble TRAIL protein. The results show that TRAIL-sensitive prostate tumor cell targets infected with Ad5-TRAIL undergo apoptosis through the production and expression of TRAIL protein. This activity was limited to TRAIL-sensitive tumor cells, as normal prostate epithelial cells were not killed by Ad5-TRAIL. Furthermore, in vivo administration of Ad5-TRAIL at the site of tumor implantation suppressed the outgrowth of human prostate tumor xenografts in SCID mice. Histologic examination of prostate tumors treated locally with Ad5-TRAIL revealed areas of apoptosis within 24 hours of injection. These results further define Ad5-TRAIL as a novel anti-tumor therapeutic and demonstrate its potential use as a means for treating prostate tumors, as well as other solid tumors, in vivo.


Subject(s)
Adenoviridae/genetics , Apoptosis , Genetic Therapy/methods , Membrane Glycoproteins/genetics , Membrane Glycoproteins/therapeutic use , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Apoptosis Regulatory Proteins , Genetic Vectors/genetics , Humans , Injections, Intralesional , Male , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/metabolism , Mice , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand , Time Factors , Transgenes/genetics , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
6.
J Immunol ; 167(6): 3049-56, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11544288

ABSTRACT

Fas ligand (FasL, CD95L) expression helps control inflammatory reactions in immune privileged sites such as the eye. Cellular activation is normally required to render lymphoid cells sensitive to FasL-induced death; however, both activated and freshly isolated Fas(+) lymphoid cells are efficiently killed in the eye. Thus, we examined factors that might regulate cell death in the eye. TNF levels rapidly increased in the eye after the injection of lymphoid cells, and these cells underwent apoptosis within 24 h. Coinjection of anti-TNF Ab with the lymphoid cells blocked this cell death. Furthermore, TNFR2(-/-) T cells did not undergo apoptosis in the eyes of normal mice, while normal and TNFR1(-/-) T cells were killed by apoptosis. In vitro, TNF enhanced the Fas-mediated apoptosis of unactivated T cells through decreased intracellular levels of FLIP and increased production of the pro-apoptotic molecule Bax. This effect was mediated through the TNFR2 receptor. In vivo, intracameral injection of normal or TNFR1(-/-) 2,4,6-trinitrophenyl-coupled T cells into normal mice induced immune deviation, but TNFR2(-/-) 2,4,6-trinitrophenyl-coupled T cells were ineffective. Collectively, our results provide evidence of a role for the p75 TNFR in cell death in that TNF signaling through TNFR2 sensitizes lymphoid cells for Fas-mediated apoptosis. We conclude that there is complicity between apoptosis and elements of the inflammatory response in controlling lymphocyte function in immune privileged sites.


Subject(s)
Anterior Chamber/immunology , Apoptosis/physiology , Eye Proteins/physiology , Graft Rejection/immunology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/physiology , Proto-Oncogene Proteins c-bcl-2 , T-Lymphocyte Subsets/cytology , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Apoptosis/drug effects , Blood-Retinal Barrier , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/physiology , Eye Proteins/pharmacology , Fas Ligand Protein , Haptens , Lymphocytes/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Picryl Chloride , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II , T-Lymphocyte Subsets/transplantation , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/pharmacology , bcl-2-Associated X Protein
7.
J Natl Cancer Inst ; 93(13): 998-1007, 2001 Jul 04.
Article in English | MEDLINE | ID: mdl-11438565

ABSTRACT

BACKGROUND: Immunization with modified tumor cells carrying recombinant immunomodulatory genes is being explored as cancer immunotherapy. In this study, we examine whether canarypox ALVAC viruses carrying immunostimulatory cytokine genes (granulocyte-macrophage colony-stimulating factor, interleukin 2, interleukin 12, and tumor necrosis factor-alpha) can induce antitumor immunity (to rechallenge) in the RM-1 model of a highly aggressive, weakly immunogenic murine prostate cancer. METHODS: For antitumor activity studies, RM-1 murine prostate cancer cells were infected with the parental ALVAC virus or one or two recombinant ALVAC-cytokine viruses and then injected into male C57BL/6 mice. For rechallenge studies, other mice were first given an injection subcutaneously with irradiated (nonproliferating) recombinant ALVAC-infected RM-1 cells and then (10 days later) with untreated RM-1 cells. For the determination of which immune cells were required for antitumor activity, mice were immunodepleted of CD4, CD8, or natural killer (NK) NK1.1 cells with the corresponding monoclonal antibodies and were then given an injection of ALVAC-cytokine-infected RM-1 cells. For all experiments, tumor outgrowth and animal survival were monitored. RESULTS: After subcutaneous injection into mice, RM-1 cells infected with one (except ALVAC-interleukin 2) or two ALVAC-cytokine recombinants had statistically significantly greater antitumor activity than RM-1 cells infected with parental ALVAC (P<.001 for all; two-sided test). The antitumor activity of RM-1 cells infected with any two ALVAC-cytokine recombinants was greater than, but not statistically significantly different from, that of RM-1 cells infected with any one ALVAC-cytokine recombinant. NK1.1 cells were necessary for antitumor activity, but tumor-specific CD4(+) regulatory T cells were also induced that inhibited CD8(+) RM-1-specific cytotoxic T cells, resulting in the lack of immunity to a rechallenge by RM-1 cells. DISCUSSION: Canarypox viruses can transfer immunostimulatory cytokine genes into RM-1 prostate cancer cells. When such cells were injected into mice, the cytokines induced an antitumor response against this highly aggressive, weakly immunogenic tumor. This response, however, did not protect the mouse against a rechallenge with RM-1 cells because suppressor CD4(+) T cells were induced that inhibited tumor-specific CD8(+) cytotoxic T cells.


Subject(s)
Avipoxvirus/genetics , Prostatic Neoplasms/therapy , Proteins , Animals , Antibodies, Monoclonal/metabolism , Antigens/biosynthesis , Antigens, Ly , Antigens, Surface , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Flow Cytometry , Gene Transfer Techniques , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-12/genetics , Interleukin-2/genetics , Lectins, C-Type , Male , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B , Neoplasm Transplantation , Prostatic Neoplasms/immunology , Protein Biosynthesis , Recombinant Proteins/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
8.
J Immunol ; 165(5): 2886-94, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10946322

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily of cytokines that induces apoptosis in a variety of cancer cells. The results presented in this study demonstrate that introduction of the human TRAIL gene into TRAIL-sensitive tumor cells using an adenoviral vector leads to the rapid production and expression of TRAIL protein, and subsequent death of the tumor cells. Tumor cell death was mediated by an apoptotic mechanism, as evidenced by the activation of caspase-8, cleavage of poly(ADP-ribose) polymerase, binding of annexin V, and inhibition by caspase inhibitor zVAD-fmk. These results define a novel method of using TRAIL as an antitumor therapeutic, and suggest the potential use for an adenovirus-encoding TRAIL as a method of gene therapy for numerous cancer types in vivo.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Apoptosis/genetics , Apoptosis/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Adenoviruses, Human/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Breast Neoplasms , Brefeldin A/pharmacology , Cell Death/drug effects , Cell Death/genetics , Cell Death/immunology , Cell Line , Cytotoxicity Tests, Immunologic , Disease Susceptibility , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/chemical synthesis , Genetic Vectors/immunology , Humans , Immunoglobulin Fc Fragments/pharmacology , Ligands , Male , Melanoma , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/pharmacology , Prostatic Neoplasms , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured/virology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Urinary Bladder Neoplasms , fas Receptor/immunology , fas Receptor/metabolism
9.
J Exp Med ; 190(8): 1155-64, 1999 Oct 18.
Article in English | MEDLINE | ID: mdl-10523613

ABSTRACT

TRAIL (TNF-related apoptosis-inducing ligand) is a member of the TNF family that induces apoptosis in a variety of cancer cells. In this study, we demonstrate that human CD11c(+) blood dendritic cells (DCs) express TRAIL after stimulation with either interferon (IFN)-gamma or -alpha and acquire the ability to kill TRAIL-sensitive tumor cell targets but not TRAIL-resistant tumor cells or normal cell types. The DC-mediated apoptosis was TRAIL specific, as soluble TRAIL receptor blocked target cell death. Moreover, IFN-stimulated interleukin (IL)-3 receptor (R)alpha(+) blood precursor (pre-)DCs displayed minimal cytotoxicity toward the same target cells, demonstrating a clear functional difference between the CD11c(+) DC and IL-3Ralpha(+) pre-DC subsets. These results indicate that TRAIL may serve as an innate effector molecule on CD11c(+) DCs for the elimination of spontaneously arising tumor cells and suggest a means by which TRAIL-expressing DCs may regulate or eliminate T cells responding to antigen presented by the DCs.


Subject(s)
Apoptosis/drug effects , Dendritic Cells/metabolism , Membrane Glycoproteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antigen Presentation/immunology , Antigens, CD/immunology , Apoptosis Regulatory Proteins , Cytokines/pharmacology , Cytotoxicity, Immunologic , Flow Cytometry , Histocytochemistry , Humans , Integrin alphaXbeta2/immunology , Interferons/pharmacology , Receptors, Interleukin-3/metabolism , TNF-Related Apoptosis-Inducing Ligand
10.
J Immunol ; 162(9): 5205-11, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10227994

ABSTRACT

Activation-induced cell death of peripheral T cells results from the interaction between Fas and Fas ligand. Resting peripheral T cells are resistant to Fas-induced apoptosis and become susceptible only after their activation. We have investigated the molecular mechanism mediating the sensitization of resting peripheral T cells to Fas-mediated apoptosis following TCR stimulation. TCR activation decreases the steady state protein levels of FLIP (FLICE-like inhibitory protein), an inhibitor of the Fas signaling pathway. Reconstitution of intracellular FLIP levels by the addition of a soluble HIV transactivator protein-FLIP chimera completely restores resistance to Fas-mediated apoptosis in TCR primary T cells. Inhibition of IL-2 production by cyclosporin A, or inhibition of IL-2 signaling by rapamycin or anti-IL-2 neutralizing Abs prevents the decrease in FLIP levels and confers resistance to Fas-mediated apoptosis following T cell activation. Using cell cycle-blocking agents, we demonstrate that activated T cells arrested in G1 phase contain high levels of FLIP protein, whereas activated T cells arrested in S phase have decreased FLIP protein levels. These findings link regulation of FLIP protein levels with cell cycle progression and provide an explanation for the increase in TCR-induced apoptosis observed during the S phase of the cell cycle.


Subject(s)
Apoptosis/immunology , Carrier Proteins/metabolism , Cell Cycle/immunology , Intracellular Signaling Peptides and Proteins , fas Receptor/physiology , Amino Acid Sequence , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/antagonists & inhibitors , Cells, Cultured , Cyclosporine/pharmacology , Down-Regulation/drug effects , Down-Regulation/immunology , Humans , Immune Sera/pharmacology , Immunity, Innate , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Molecular Sequence Data , Receptors, Antigen, T-Cell/physiology , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
11.
J Exp Med ; 189(8): 1343-54, 1999 Apr 19.
Article in English | MEDLINE | ID: mdl-10209050

ABSTRACT

TRAIL (tumor necrosis factor [TNF]-related apoptosis-inducing ligand) is a molecule that displays potent antitumor activity against selected targets. The results presented here demonstrate that human monocytes rapidly express TRAIL, but not Fas ligand or TNF, after activation with interferon (IFN)-gamma or -alpha and acquire the ability to kill tumor cells. Monocyte-mediated tumor cell apoptosis was TRAIL specific, as it could be inhibited with soluble TRAIL receptor. Moreover, IFN stimulation caused a concomitant loss of TRAIL receptor 2 expression, which coincides with monocyte acquisition of resistance to TRAIL-mediated apoptosis. These results define a novel mechanism of monocyte-induced cell cytotoxicity that requires TRAIL, and suggest that TRAIL is a key effector molecule in antitumor activity in vivo.


Subject(s)
Antineoplastic Agents/metabolism , Membrane Glycoproteins/metabolism , Monocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/pharmacology , Phosphatidylserines/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
12.
J Immunol ; 162(5): 2597-605, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10072501

ABSTRACT

mAbs were generated against the extracellular domain of the four known TNF-related apoptosis-inducing ligand (TRAIL) receptors and tested on a panel of human melanoma cell lines. The specificity of the mAb permitted a precise evaluation of the TRAIL receptors that induce apoptosis (TRAIL-R1 and -R2) compared with the TRAIL receptors that potentially regulate TRAIL-mediated apoptosis (TRAIL-R3 and -R4). Immobilized anti-TRAIL-R1 or -R2 mAbs were cytotoxic to TRAIL-sensitive tumor cells, whereas tumor cells resistant to recombinant TRAIL were also resistant to these mAbs and only became sensitive when cultured with actinomycin D. The anti-TRAIL-R1 and -R2 mAb-induced death was characterized by the activation of intracellular caspases, which could be blocked by carbobenzyloxy-Val-Ala-Asp (OMe) fluoromethyl ketone (zVAD-fmk) and carbobenzyloxy-Ile-Glu(OMe)-Thr-Asp (OMe) fluoromethyl ketone (zIETD-fmk). When used in solution, one of the anti-TRAIL-R2 mAbs was capable of blocking leucine zipper-human TRAIL binding to TRAIL-R2-expressing cells and prevented TRAIL-induced death of these cells, whereas two of the anti-TRAIL-R1 mAbs could inhibit leucine zipper-human TRAIL binding to TRAIL-R1:Fc. Furthermore, use of the blocking anti-TRAIL-R2 mAb allowed us to demonstrate that the signals transduced through either TRAIL-R1 or TRAIL-R2 were necessary and sufficient to mediate cell death. In contrast, the expression of TRAIL-R3 or TRAIL-R4 did not appear to be a significant factor in determining the resistance or sensitivity of these tumor target cells to the effects of TRAIL.


Subject(s)
Antibodies, Monoclonal/immunology , Apoptosis , Receptors, Tumor Necrosis Factor/physiology , Animals , Apoptosis Regulatory Proteins , Caspase Inhibitors , GPI-Linked Proteins , Humans , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor, Member 10c , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor-alpha/antagonists & inhibitors
13.
Nat Med ; 5(2): 157-63, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9930862

ABSTRACT

To evaluate the utility of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) as a cancer therapeutic, we created leucine zipper (LZ) forms of human (hu) and murine (mu) TRAIL to promote and stabilize the formation of trimers. Both were biologically active, inducing apoptosis of both human and murine target cells in vitro with similar specific activities. In contrast to the fulminant hepatotoxicity of LZ-huCD95L in vivo, administration of either LZ-huTRAIL or LZ-muTRAIL did not seem toxic to normal tissues of mice. Finally, repeated treatments with LZ-huTRAIL actively suppressed growth of the TRAIL-sensitive human mammary adenocarcinoma cell line MDA-231 in CB.17 (SCID) mice, and histologic examination of tumors from SCID mice treated with LZ-huTRAIL demonstrated clear areas of apoptotic necrosis within 9-12 hours of injection.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis Regulatory Proteins , Dose-Response Relationship, Drug , Fas Ligand Protein , Humans , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/chemical synthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Protein Conformation , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/chemical synthesis
14.
Immunity ; 9(5): 711-20, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9846492

ABSTRACT

Fas (CD95) and Fas ligand (FasL) play major roles in staphylococcal enterotoxin B (SEB)-induced peripheral deletion of Vbeta8+ T cells. We found that peripheral deletion was defective in radiation chimeras with non-functional tissue FasL, regardless of the FasL status of the bone marrow-derived cells. SEB induced a dramatic upregulation of FasL expression and function in nonlymphoid cells of liver and small intestine. This effect was resistant to inhibition by cyclosporin A, which also failed to inhibit peripheral deletion. In SCID animals nonlymphoid tissues did not express FasL in response to SEB unless transplanted lymphocytes were present. Thus, some immune responses induce FasL in nonlymphoid tissues, which in turn kills activated lymphocytes, leading to peripheral T cell deletion.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Membrane Glycoproteins/biosynthesis , Superantigens/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cyclosporine/pharmacology , Enterotoxins/pharmacology , Fas Ligand Protein , Immunosuppressive Agents/pharmacology , In Situ Hybridization , Intestine, Small/cytology , Intestine, Small/metabolism , Liver/cytology , Liver/metabolism , Lymphocyte Activation/drug effects , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, SCID , Radiation Chimera/immunology , Reverse Transcriptase Polymerase Chain Reaction , Superantigens/immunology
15.
Curr Opin Immunol ; 10(5): 559-63, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9794836

ABSTRACT

Apoptosis research is benefiting from bioinformatic approaches to identify new components of the cell death machinery and novel cell death inducers/receptors. Over the past year, knowledge of the system involving TNF-related apoptosis-inducing ligand (TRAIL) and its receptors has increased via genomic database analysis to include four distinct receptors that interact with a single ligand. Currently, these molecules are of major interest due to their potential roles and application in cancer therapy.


Subject(s)
Apoptosis , Membrane Glycoproteins/physiology , Receptors, Tumor Necrosis Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , GPI-Linked Proteins , Humans , RNA, Messenger/analysis , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Member 10c , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor Decoy Receptors
16.
J Exp Med ; 188(5): 887-96, 1998 Sep 07.
Article in English | MEDLINE | ID: mdl-9730890

ABSTRACT

Apoptosis is critical to homeostasis of multicellular organisms. In immune privileged sites such as the eye, CD95 ligand (FasL)-induced apoptosis controls dangerous inflammatory reactions that can cause blindness. Recently, we demonstrated that apoptotic cell death of inflammatory cells was a prerequisite for the induction of immune deviation after antigen presentation in the eye. In this report, we examine the mechanism by which this takes place. Our results show that Fas- mediated apoptosis of lymphoid cells leads to rapid production of interleukin (IL)-10 in these cells. The apoptotic cells containing IL-10 are responsible for the activation of immune deviation through interaction with antigen-presenting cells (APC). In support of this, we found that apoptotic cells from IL-10(+/+) animals fed to APC in vitro promote Th2 cell differentiation, whereas apoptotic IL-10(-/-) cells, as well as nonapoptotic cells, favor Th1 induction. Thus, apoptotic cell death and tolerance are linked through the production of an antiinflammatory cytokine to prevent dangerous and unwanted immune responses that might compromise organ integrity.


Subject(s)
Apoptosis/immunology , Membrane Glycoproteins/physiology , fas Receptor/physiology , Animals , Anterior Chamber/immunology , Anterior Chamber/metabolism , Antigen Presentation , Cell Differentiation/immunology , Fas Ligand Protein , Inflammation/immunology , Inflammation/prevention & control , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-4/genetics , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Th2 Cells/cytology , Th2 Cells/metabolism
17.
J Immunol ; 161(6): 2833-40, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9743343

ABSTRACT

The observation that TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF cytokine family, induces apoptosis in a number of different tumor cell types led us to compare the tumoricidal effects of TRAIL to those of other TNF family molecules on human melanoma cells. We found that a high proportion of the melanoma cell lines tested were killed by TRAIL, whereas all the melanoma lines were resistant to the other TNF family cytokines tested. TRAIL-induced death was characterized by caspase activation and cellular protein cleavage within minutes of TRAIL addition, and death could be completely inhibited by the caspase inhibitors Ile-Glu-Thr-Asp (IETD) and Val-Ala-Asp (VAD), indicating the presence of a TRAIL receptor signaling pathway similar to that identified for Fas and TNF receptors. Specific TRAIL receptor expression was determined by RT-PCR, and the presence of mRNA encoding the "protective" TRAIL receptors did not correspond to resistance or sensitivity to TRAIL-induced apoptosis. Addition of protein synthesis inhibitors to TRAIL-resistant melanomas rendered them sensitive to TRAIL, indicating that the presence or the absence of intracellular apoptosis inhibitors may mediate resistance or sensitivity to TRAIL-mediated apoptosis. Expression of one such inhibitor, FLICE-inhibitory protein (FLIP), was highest in the TRAIL-resistant melanomas, while being low or undetectable in the TRAIL-sensitive melanomas. Furthermore, addition of actinomycin D to TRAIL-resistant melanomas resulted in decreased intracellular concentrations of FLIP, which correlated with their acquisition of TRAIL sensitivity. Collectively, our results indicate that TRAIL-induced apoptosis occurs through a caspase signaling cascade and that resistance is controlled by intracellular regulators of apoptosis.


Subject(s)
Apoptosis/immunology , Intracellular Fluid/metabolism , Intracellular Signaling Peptides and Proteins , Melanoma/pathology , Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , Amino Acid Sequence , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/physiology , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/physiology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Humans , Immunity, Innate , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Ligands , Melanoma/enzymology , Melanoma/immunology , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , Protein Synthesis Inhibitors/pharmacology , Receptors, Tumor Necrosis Factor/biosynthesis , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
19.
Immunol Rev ; 156: 167-84, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9176707

ABSTRACT

Immune privilege is a term applied to several organs that have a unique relationship with the immune response. These sites prohibit the spread of inflammation since even minor episodes can threaten organ integrity and function. The most prominent examples of these are the eye, brain and reproductive organs where immune responses either do not proceed, or proceed in a manner different from other areas. Once thought to be a passive process relying on physical barriers, immune privilege can now be viewed as an active process that utilizes multiple mechanisms to maintain organ function. Recently there has been a renewed interest in immune privilege when it was shown that two privileged sites (the eye and testes) constitutively express FasL, which functions by killing lymphoid cells that invade these areas. Here we will examine the role of FasL in immune privilege and discuss how this molecule interacts with other elements of the inflammatory response to maintain organ integrity in the face of potentially damaging immune reactions.


Subject(s)
Cell Death , Eye/immunology , Membrane Glycoproteins/immunology , fas Receptor/immunology , Animals , Cell Adhesion , Eye/cytology , Eye/ultrastructure , Fas Ligand Protein , Forecasting , Humans , Lymphoid Tissue , Neuropeptides/immunology , Substance P/immunology , Transplantation Immunology , Vasoactive Intestinal Peptide/immunology
20.
J Clin Invest ; 99(3): 396-402, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9022072

ABSTRACT

Although anatomical barriers and soluble mediators have been implicated in immune privilege, it appears that the apoptotic cell death of Fas+ cells by tissue-associated CD95 ligand (Fas ligand, FasL) is an important component. One clinical example of the function of an immune privileged site is the success of human corneal transplants, where a very high percentage of transplants accept without tissue matching or immunosuppressive therapy. Since the mouse cornea expresses abundant Fas ligand and immune privilege has been implicated in the success of these transplants, we examined the role of FasL in corneal transplantation. Our results show that human corneas express functional FasL capable of killing Fas+ lymphoid cells in an in vitro culture system. Using a mouse model for corneal allograft transplantation, FasL+ orthografts were accepted at a rate of 45%, whereas FasL- grafts, or normal grafts transplanted to Fas- mice, were rejected 100% of the time. Histological analysis found that FasL+ grafts contained apoptotic mononuclear cells indicating the induction of apoptosis by the graft, while rejecting FasL- corneas contained numerous inflammatory cells without associated apoptosis. Taken together our results demonstrate that FasL expression on the cornea is a major factor in corneal allograft survival and, thus, we provide an explanation for one of the most successful tissue transplants performed in humans.


Subject(s)
Corneal Transplantation/immunology , Graft Survival/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/physiology , Transplantation, Homologous/immunology , fas Receptor/immunology , Actins/genetics , Adolescent , Adult , Aged , Animals , Apoptosis/immunology , Child , Child, Preschool , Cornea/immunology , Cytotoxicity, Immunologic , Fas Ligand Protein , Female , Graft Rejection/immunology , HeLa Cells , Histocompatibility Testing , Humans , Immunohistochemistry , Immunosuppression Therapy , Infant , Infant, Newborn , Inflammation , Male , Membrane Glycoproteins/genetics , Mice , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...