Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
J Biol Chem ; 276(15): 12060-7, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11278459

ABSTRACT

Cytotoxic T lymphocytes kill virus-infected and tumor cell targets through the concerted action of proteins contained in cytolytic granules, primarily granzyme B and perforin. Granzyme B, a serine proteinase with substrate specificity similar to the caspase family of apoptotic cysteine proteinases, is capable of cleaving and activating a number of death proteins in target cells. Despite the ability to engage the death pathway at multiple entry points, the preferred mechanism for rapid induction of apoptosis by granzyme B has yet to be clearly established. Here we use time lapse confocal microscopy to demonstrate that mitochondrial cytochrome c release is the primary mode of granzyme B-induced apoptosis and that Bcl-2 is a potent inhibitor of this pivotal event. Caspase activation is not required for cytochrome c release, an activity that correlates with cleavage and activation of Bid, which we have found to be cleaved more readily by granzyme B than either caspase-3 or caspase-8. Bcl-2 blocks the rapid destruction of targets by granzyme B by blocking mitochondrial involvement in the process.


Subject(s)
Apoptosis/drug effects , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Serine Endopeptidases/pharmacology , Amino Acid Sequence , Cytochrome c Group/metabolism , Enzyme Activation , Granzymes , Humans , Hydrolysis , Jurkat Cells , Kinetics , Molecular Sequence Data
3.
J Exp Med ; 192(10): 1391-402, 2000 Nov 20.
Article in English | MEDLINE | ID: mdl-11085742

ABSTRACT

Cytotoxic T lymphocytes (CTLs) destroy target cells through a mechanism involving the exocytosis of cytolytic granule components including granzyme B (grB) and perforin, which have been shown to induce apoptosis through caspase activation. However, grB has also been linked with caspase-independent disruption of mitochondrial function. We show here that cytochrome c release requires the direct proteolytic cleavage of Bid by grB to generate a 14-kD grB-truncated product (gtBid) that translocates to mitochondria. In turn, gtBid recruits Bax to mitochondria through a caspase-independent mechanism where it becomes integrated into the membrane and induces cytochrome c release. Our results provide evidence for a new pathway by which CTLs inflict damage and explain the caspase-independent mechanism of mitochondrial dysfunction.


Subject(s)
Carrier Proteins/metabolism , Cytochrome c Group/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Serine Endopeptidases/metabolism , BH3 Interacting Domain Death Agonist Protein , Cell Death , Cytosol/metabolism , Cytotoxicity, Immunologic , Granzymes , Humans , Intracellular Membranes/metabolism , Jurkat Cells/virology , Models, Biological , Protein Processing, Post-Translational , Protein Transport , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured , bcl-2-Associated X Protein
4.
Cell ; 103(3): 491-500, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-11081635

ABSTRACT

The serine proteinase granzyme B is crucial for the rapid induction of target cell apoptosis by cytotoxic T cells. Granzyme B was recently demonstrated to enter cells in a perforin-independent manner, thus predicting the existence of a cell surface receptor(s). We now present evidence that this receptor is the cation-independent mannose 6-phosphate/insulin-like growth factor receptor (CI-MPR). Inhibition of the granzyme B-CI-MPR interaction prevented granzyme B cell surface binding, uptake, and the induction of apoptosis. Significantly, expression of the CI-MPR was essential for cytotoxic T cell-mediated apoptosis of target cells in vitro and for the rejection of allogeneic cells in vivo. These results suggest a novel target for immunotherapy and a potential mechanism used by tumors for immune evasion.


Subject(s)
Apoptosis/drug effects , Receptor, IGF Type 2/metabolism , Serine Endopeptidases/metabolism , Serine Endopeptidases/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Animals , Binding, Competitive/drug effects , Cell Transplantation , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Endocytosis/drug effects , Flow Cytometry , Graft Rejection/immunology , Graft Rejection/metabolism , Granzymes , Humans , In Situ Nick-End Labeling , Jurkat Cells , Kidney/immunology , L Cells , Mannosephosphates/metabolism , Mannosephosphates/pharmacology , Mice , Mice, Inbred BALB C , Mice, SCID , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Binding/drug effects , Receptor, IGF Type 2/antagonists & inhibitors
5.
Mol Cell Biol ; 20(11): 3781-94, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10805722

ABSTRACT

Cytotoxic T lymphocytes (CTL) can trigger an apoptotic signal through the Fas receptor or by the exocytosis of granzyme B and perforin. Caspase activation is an important component of both pathways. Granzyme B, a serine proteinase contained in granules, has been shown to proteolytically process and activate members of the caspase family in vitro. In order to gain an understanding of the contributions of caspases 8 and 3 during granule-induced apoptosis in intact cells, we have used target cells that either stably express the rabbitpox virus-encoded caspase inhibitor SPI-2 or are devoid of caspase 3. The overexpression of SPI-2 in target cells significantly inhibited DNA fragmentation, phosphatidylserine externalization, and mitochondrial disruption during Fas-mediated cell death. In contrast, SPI-2 expression in target cells provided no protection against granzyme-mediated apoptosis, mitochondrial collapse, or cytolysis, leading us to conclude that SPI-2-inhibited caspases are not an essential requirement for the granzyme pathway. Caspase 3-deficient MCF-7 cells were found to be resistant to CTL-mediated DNA fragmentation but not to CTL-mediated cytolysis and loss of the mitochondrial inner membrane potential. Furthermore, we demonstrate that granzyme B directly cleaves the proapoptotic molecule Bid, bypassing the need for caspase 8 activation of Bid. These results provide evidence for a two-pronged strategy for mediating target cell destruction and provide evidence of a direct link between granzyme B activity, Bid cleavage, and caspase 3 activation in whole cells.


Subject(s)
Apoptosis/immunology , Carrier Proteins/metabolism , Caspases/metabolism , Serine Endopeptidases/metabolism , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins , BH3 Interacting Domain Death Agonist Protein , Caspase 8 , Caspase 9 , Cysteine Proteinase Inhibitors/biosynthesis , Cysteine Proteinase Inhibitors/genetics , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic/immunology , DNA Fragmentation , Enzyme Activation , Gene Expression , Granzymes , Humans , Jurkat Cells , Serpins/biosynthesis , Serpins/genetics , Tumor Cells, Cultured , fas Receptor/immunology
6.
Cell Death Differ ; 7(1): 17-24, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10713717

ABSTRACT

Granzyme B is the prototypic member of a family of serine proteases localized to the cytolytic granules of cytotoxic lymphocytes. Together with another granule protein, perforin, granzyme B is capable of inducing all aspects of apoptotic death in target cells. A number of granzyme B substrates have been identified and it has been demonstrated that granzyme B is responsible, directly or indirectly, for the morphological nuclear changes observed in target cell apoptosis, including DNA fragmentation. In an earlier study, we showed that granzyme B binds to a nuclear protein in a manner dependent on its enzymatic activity. Here, we demonstrate that granzyme B is translocated rapidly to the nucleus in cells that have been induced to undergo apoptosis by a granzyme-dependent process, and that translocation is dependent on caspase activity. Appearance of granzyme B in the nucleus of target cells precedes the detection of DNA fragmentation. Although not directly responsible for DNA fragmentation, these data suggest a nuclear role for granzyme B in target cell apoptosis. c-Abl nuclear functions.


Subject(s)
Apoptosis , Serine Endopeptidases/metabolism , Animals , Apoptosis/drug effects , Biological Transport , COS Cells , Caspase 3 , Caspases/metabolism , Cell Membrane/physiology , Cell Nucleus/metabolism , DNA Fragmentation , Enzyme Activation , Granzymes , Humans , Jurkat Cells
7.
J Immunol ; 163(9): 4683-93, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10528165

ABSTRACT

CTLs kill targets by inducing them to die through apoptosis. A number of morphological and biochemical events are now recognized as characteristic features of the apoptotic program. Among these, the disruption of the inner mitochondrial transmembrane potential (Delta Psi m) and the release of cytochrome c into the cytoplasm appear to be early events in many systems, leading to the activation of caspase-3 and, subsequently, nuclear apoptosis. We show here that, in Jurkat targets treated in vitro with purified granzyme B and perforin or granzyme B and adenovirus, Delta Psi m collapse, reactive oxygen species production, and cytochrome c release from mitochondria were observed. Loss of Delta Psi m was also detected in an in vivo system where green fluorescent protein-expressing targets were attacked by a cytotoxic T cell line that kills predominantly through the granzyme pathway. DNA fragmentation, phosphatidylserine externalization, and reactive oxygen species production were inhibited in the presence of the caspase inhibitors benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (zVAD-fmk) and benzyloxycarbonyl-Asp-Glu-Val-Asp-fluoromethyl ketone (zDEVD-fmk) in our in vitro system. Importantly, in either the in vitro or in vivo systems, these inhibitors at concentrations up to 100 microM did not prevent Delta Psi m collapse. In addition, cytochrome c release was observed in the in vitro system in the absence or presence of zVAD-fmk. Thus the granzyme B-dependent killing pathway in Jurkat targets involves mitochondrial alterations that occur independently of caspases.


Subject(s)
Caspases/metabolism , Cytochrome c Group/metabolism , Intracellular Membranes/enzymology , Mitochondria/enzymology , Serine Endopeptidases/physiology , Adenoviruses, Human/immunology , Animals , Apoptosis/immunology , Cyclosporine/pharmacology , DNA Fragmentation/immunology , Enzyme Activation/drug effects , Granzymes , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Membranes/pathology , Jurkat Cells , Membrane Glycoproteins/immunology , Membrane Potentials/drug effects , Membrane Potentials/immunology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Perforin , Phosphatidylserines/metabolism , Pore Forming Cytotoxic Proteins , Rats , Reactive Oxygen Species/metabolism , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
8.
Blood ; 92(3): 1044-54, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9680374

ABSTRACT

In the widely accepted model of granule-mediated killing by cytotoxic lymphocytes, granzyme B entry into the target cell is facilitated by the pore forming molecule, perforin. Using indirect immunofluorescence and also direct visualization of fluorescein isothiocyanate (FITC)-conjugated granzyme B, we demonstrate internalization in the absence of perforin. Induction of the lytic pathway, however, required a second signal that was provided by perforin or adenovirus (Ad2). The combination of agents also resulted in a dramatic relocalization of the granzyme. Microinjection of granzyme B directly into the cytoplasm of target cells resulted in apoptosis without the necessity of a second stimulus. This suggested that the key event is the presence of granzyme B in the cytoplasm, and that when the enzyme is internalized by a target cell, it trafficks to an intracellular compartment and accumulates until release is stimulated by the addition of perforin. We found that the proteinase passed through rab5-positive vesicles and then accumulated within a novel compartment. On the basis of these results, we propose a new model for granzyme-perforin-induced target cell lysis in which granzyme B is subjected to trafficking events in the target cell that control and contribute to cell death.


Subject(s)
Apoptosis/physiology , Endocytosis , Membrane Glycoproteins/physiology , Serine Endopeptidases/physiology , T-Lymphocytes, Cytotoxic/metabolism , Adenoviridae/physiology , Animals , Biological Transport , COS Cells/drug effects , COS Cells/metabolism , Cell Compartmentation , Cell Nucleus/metabolism , Cytoplasm/metabolism , Endosomes/metabolism , GTP-Binding Proteins/physiology , Granzymes , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , Jurkat Cells/cytology , Jurkat Cells/metabolism , Membrane Glycoproteins/pharmacology , Microinjections , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Perforin , Pore Forming Cytotoxic Proteins , Recombinant Proteins/metabolism , Serine Endopeptidases/pharmacology , T-Lymphocytes, Cytotoxic/cytology , Transfection , Tumor Cells, Cultured , rab5 GTP-Binding Proteins
9.
J Exp Med ; 178(6): 1935-46, 1993 Dec 01.
Article in English | MEDLINE | ID: mdl-8245774

ABSTRACT

The intracellular distribution of the enzyme 5-lipoxygenase (5-LO) and 5-lipoxygenase-activating protein (FLAP) in resting and ionophore-activated human leukocytes has been determined using immuno-electronmicroscopic labeling of ultrathin frozen sections and subcellular fractionation techniques. 5-LO is a 78-kD protein that catalyzes the conversion of arachidonic acid to leukotrienes. FLAP is an 18-kD membrane bound protein that is essential for leukotriene synthesis in cells. In response to ionophore stimulation, 5-LO translocates from a soluble to a sedimentable fraction of cell homogenates. In activated leukocytes, both FLAP and 5-LO were localized in the lumen of the nuclear envelope. Neither protein could be detected in any other cell compartment or along the plasma membrane. In resting cells, the FLAP distribution was identical to that observed in activated cells. In addition, subcellular fractionation techniques showed > 83% of immunoblot-detectable FLAP protein and approximately 64% of the FLAP ligand binding activity was found in the nuclear membrane fraction. A fractionation control demonstrated that a plasma membrane marker, detected by a monoclonal antibody PMN13F6, was not detectable in the nuclear membrane fraction. In contrast to FLAP, 5-LO in resting cells could not be visualized along the nuclear envelope. Except for weak labeling of the euchromatin region of the nucleus, 5-LO could not be readily detected in any other cellular compartment. These results demonstrate that the nuclear envelope is the intracellular site at which 5-LO and FLAP act to metabolize arachidonic acid, and that ionophore activation of neutrophils and monocytes results in the translocation of 5-LO from a nonsedimentable location to the nuclear envelope.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Monocytes/enzymology , Neutrophils/enzymology , Nuclear Envelope/enzymology , 5-Lipoxygenase-Activating Proteins , Blotting, Western , Calcimycin/pharmacology , Cell Compartmentation , Humans , Immunohistochemistry , In Vitro Techniques , Monocytes/ultrastructure , Neutrophils/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...