Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Trends Biochem Sci ; 39(11): 510-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25440714

ABSTRACT

Pore-forming proteins (PFPs) interact with lipid bilayers to compromise membrane integrity. Many PFPs function by inserting a ring of oligomerized subunits into the bilayer to form a protein-lined hydrophilic channel. However, mounting evidence suggests that PFPs can also generate 'proteolipidic' pores by contributing to the fusion of inner and outer bilayer leaflets to form a toroidal structure. We discuss here toroidal pore formation by peptides including melittin, protegrin, and Alzheimer's Aß1-41, as well as by PFPs from several evolutionarily unrelated families: the colicin/Bcl-2 grouping including the pro-apoptotic protein Bax, actinoporins derived from sea anemones, and the membrane attack complex-perforin/cholesterol dependent cytolysin (MACPF/CDC) set of proteins. We also explore how the structure and biological role of toroidal pores might be investigated further.


Subject(s)
Cell Membrane/chemistry , Lipid Bilayers/chemistry , Membrane Lipids/chemistry , Pore Forming Cytotoxic Proteins/chemistry , Cell Membrane/metabolism , Colicins/chemistry , Colicins/metabolism , Lipid Bilayers/metabolism , Melitten/chemistry , Melitten/metabolism , Membrane Lipids/metabolism , Models, Molecular , Pore Forming Cytotoxic Proteins/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
2.
Immunity ; 29(5): 720-33, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18951048

ABSTRACT

Granzyme A (GzmA) is considered a major proapoptotic protease. We have discovered that GzmA-induced cell death involves rapid membrane damage that depends on the synergy between micromolar concentrations of GzmA and sublytic perforin (PFN). Ironically, GzmA and GzmB, independent of their catalytic activity, both mediated this swift necrosis. Even without PFN, lower concentrations of human GzmA stimulated monocytic cells to secrete proinflammatory cytokines (interleukin-1beta [IL-1beta], TNFalpha, and IL-6) that were blocked by a caspase-1 inhibitor. Moreover, murine GzmA and GzmA(+) cytotoxic T lymphocytes (CTLs) induce IL-1beta from primary mouse macrophages, and GzmA(-/-) mice resist lipopolysaccharide-induced toxicity. Thus, the granule secretory pathway plays an unexpected role in inflammation, with GzmA acting as an endogenous modulator.


Subject(s)
Granzymes/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Leukocytes, Mononuclear/immunology , Perforin/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/immunology , Adenoviridae/immunology , Animals , Cell Adhesion , Cell Death , Cell Line, Tumor , Cytotoxicity, Immunologic , Gene Knockdown Techniques , Granzymes/metabolism , HeLa Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Jurkat Cells , Macrophages/immunology , Mice , Perforin/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Tumor Necrosis Factor-alpha/metabolism , U937 Cells
3.
J Biol Chem ; 290(11): 6868-77, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25605735

ABSTRACT

Cytotoxic T cells (Tc) use perforin and granzyme B (gzmB) to kill virus-infected cells and cancer cells. Recent evidence suggests that human gzmB primarily induces apoptosis via the intrinsic mitochondrial pathway by either cleaving Bid or activating Bim leading to the activation of Bak/Bax and subsequent generation of active caspase-3. In contrast, mouse gzmB is thought to predominantly induce apoptosis by directly processing pro-caspase-3. However, in certain mouse cell types gzmB-mediated apoptosis mainly occurs via the mitochondrial pathway. To investigate whether Bim is involved under the latter conditions, we have now employed ex vivo virus-immune mouse Tc that selectively kill by using perforin and gzmB (gzmB(+)Tc) as effector cells and wild type as well as Bim- or Bak/Bax-deficient spontaneously (3T9) or virus-(SV40) transformed mouse embryonic fibroblast cells as targets. We show that gzmB(+)Tc-mediated apoptosis (phosphatidylserine translocation, mitochondrial depolarization, cytochrome c release, and caspase-3 activation) was severely reduced in 3T9 cells lacking either Bim or both Bak and Bax. This outcome was related to the ability of Tc cells to induce the degradation of Mcl-1 and Bcl-XL, the anti-apoptotic counterparts of Bim. In contrast, gzmB(+)Tc-mediated apoptosis was not affected in SV40-transformed mouse embryonic fibroblast cells lacking Bak/Bax. The data provide evidence that Bim participates in mouse gzmB(+)Tc-mediated apoptosis of certain targets by activating the mitochondrial pathway and suggest that the mode of cell death depends on the target cell. Our results suggest that the various molecular events leading to transformation and/or immortalization of cells have an impact on their relative resistance to the multiple gzmB(+)Tc-induced death pathways.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Apoptosis , Granzymes/immunology , Membrane Proteins/immunology , Mitochondria/immunology , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Cytotoxic/enzymology , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspase 3/immunology , Cell Line , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Deletion , Immunotherapy , Membrane Proteins/genetics , Mice , Myeloid Cell Leukemia Sequence 1 Protein/immunology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/therapy , Pore Forming Cytotoxic Proteins/immunology , Proteolysis , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology
4.
PLoS Pathog ; 9(1): e1003119, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23326234

ABSTRACT

Human γ(9)δ(2) T cells potently inhibit pathogenic microbes, including intracellular mycobacteria, but the key inhibitory mechanism(s) involved have not been identified. We report a novel mechanism involving the inhibition of intracellular mycobacteria by soluble granzyme A. γ(9)δ(2) T cells produced soluble factors that could pass through 0.45 µm membranes and inhibit intracellular mycobacteria in human monocytes cultured below transwell inserts. Neutralization of TNF-α in co-cultures of infected monocytes and γ(9)δ(2) T cells prevented inhibition, suggesting that TNF-α was the critical inhibitory factor produced by γ(9)δ(2) T cells. However, only siRNA- mediated knockdown of TNF-α in infected monocytes, but not in γ(9)δ(2) T cells, prevented mycobacterial growth inhibition. Investigations of other soluble factors produced by γ(9)δ(2) T cells identified a highly significant correlation between the levels of granzyme A produced and intracellular mycobacterial growth inhibition. Furthermore, purified granzyme A alone induced inhibition of intracellular mycobacteria, while knockdown of granzyme A in γ(9)δ(2) T cell clones blocked their inhibitory effects. The inhibitory mechanism was independent of autophagy, apoptosis, nitric oxide production, type I interferons, Fas/FasL and perforin. These results demonstrate a novel microbial defense mechanism involving granzyme A-mediated triggering of TNF-α production by monocytes leading to intracellular mycobacterial growth suppression. This pathway may provide a protective mechanism relevant for the development of new vaccines and/or immunotherapies for macrophage-resident chronic microbial infections.


Subject(s)
Granzymes/metabolism , Macrophages/enzymology , Monocytes/enzymology , Mycobacterium/physiology , T-Lymphocyte Subsets/enzymology , Cells, Cultured , Gene Expression Regulation, Bacterial , Gene Knockdown Techniques , Granzymes/genetics , Granzymes/pharmacology , Host-Pathogen Interactions , Humans , Macrophages/immunology , Macrophages/microbiology , Monocytes/immunology , Monocytes/microbiology , Mycobacterium/drug effects , Neutralization Tests , RNA, Small Interfering/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Tumor Necrosis Factor-alpha/metabolism
5.
Cell Mol Life Sci ; 70(12): 2083-98, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22983385

ABSTRACT

Recent work on the MACPF/CDC superfamily of pore-forming proteins has focused on the structural analysis of monomers and pore-forming oligomeric complexes. We set the family of proteins in context and highlight aspects of their function which the direct and exclusive equation of oligomers with pores fails to explain. Starting with a description of the distribution of MACPF/CDC proteins across the domains of life, we proceed to show how their evolutionary relationships can be understood on the basis of their structural homology and re-evaluate models for pore formation by perforin, in particular. We furthermore highlight data showing the role of incomplete oligomeric rings (arcs) in pore formation and how this can explain small pores generated by oligomers of proteins belonging to the family. We set this in the context of cell biological and biophysical data on the proteins' function and discuss how this helps in the development of an understanding of how they act in processes such as apicomplexan parasites gliding through cells and exiting from cells.


Subject(s)
Cell Membrane/metabolism , Complement Membrane Attack Complex/metabolism , Cytotoxins/metabolism , Models, Molecular , Perforin/metabolism , Phylogeny , Protein Conformation , Amino Acid Sequence , Apicomplexa , Bacteria , Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/genetics , Cytotoxins/chemistry , Molecular Sequence Data , Perforin/chemistry , Polymerization , Sequence Alignment
6.
Proc Natl Acad Sci U S A ; 108(52): 21016-21, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22173634

ABSTRACT

The cytotoxic cell granule secretory pathway is essential for immune defence. How the pore-forming protein perforin (PFN) facilitates the cytosolic delivery of granule-associated proteases (granzymes) remains enigmatic. Here we show that PFN is able to induce invaginations and formation of complete internal vesicles in giant unilamellar vesicles. Formation of internal vesicles depends on native PFN and calcium and antibody labeling shows the localization of PFN at the invaginations. This vesiculation is recapitulated in large unilamellar vesicles and in this case PFN oligomers can be seen associated with the necks of the invaginations. Capacitance measurements show PFN is able to increase a planar lipid membrane surface area in the absence of pore formation, in agreement with the ability to induce invaginations. Finally, addition of PFN to Jurkat cells causes the formation of internal vesicles prior to pore formation. PFN is capable of triggering an endocytosis-like event in addition to pore formation, suggesting a new paradigm for its role in delivering apoptosis-inducing granzymes into target cells.


Subject(s)
Cell Membrane/metabolism , Endocytosis/physiology , Granzymes/metabolism , Immunity, Innate/physiology , Perforin/metabolism , Secretory Vesicles/metabolism , Cryoelectron Microscopy , Humans , Jurkat Cells , Microscopy, Fluorescence , Perforin/immunology , Perforin/physiology
7.
J Virol ; 86(16): 8713-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22674984

ABSTRACT

The T cell granule exocytosis pathway is essential to control hepatotropic lymphocytic choriomeningitis virus strain WE (LCMV-WE) but also contributes to the observed pathology in mice. Although effective antiviral T cell immunity and development of viral hepatitis are strictly dependent on perforin and granzymes, the molecular basis underlying induction of functionally competent virus-immune T cells, including participation of the innate immune system, is far from being resolved. We demonstrate here that LCMV-immune T cells of interleukin-1 receptor (IL-1R)-deficient mice readily express transcripts for perforin and granzymes but only translate perforin, resulting in the lack of proapoptotic potential in vitro. LCMV is not cleared in IL-1R-deficient mice, and yet the infected mice develop neither splenomegaly nor hepatitis. These results demonstrate that IL-1R signaling is central to the induction of proapoptotic CD8 T cell immunity, including viral clearance and associated tissue injuries in LCMV infection.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Interleukin-1/immunology , Animals , Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Disease Models, Animal , Hepatitis/immunology , Hepatitis/pathology , Hepatitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1/deficiency , Splenomegaly/immunology , Splenomegaly/pathology , Splenomegaly/virology
8.
J Biol Chem ; 286(4): 2946-55, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-20889983

ABSTRACT

Perforin (PFN) is a pore-forming protein produced by cytotoxic lymphocytes that aids in the clearance of tumor or virus-infected cells by a mechanism that involves the formation of transmembrane pores. The properties of PFN pores and the mechanism of their assembly remain unclear. Here, we studied pore characteristics by functional and structural methods to show that perforin forms pores more heterogeneous than anticipated. Planar lipid bilayer experiments indicate that perforin pores exhibit a broad range of conductances, from 0.15 to 21 nanosiemens. In comparison with large pores that possessed low noise and remained stably open, small pores exhibited high noise and were very unstable. Furthermore, the opening step and the pore size were dependent on the lipid composition of the membrane. The heterogeneity in pore sizes was confirmed with cryo-electron microscopy and showed a range of sizes matching that observed in the conductance measurements. Furthermore, two different membrane-bound PFN conformations were observed, interpreted as pre-pore and pore states of the protein. The results collectively indicate that PFN forms heterogeneous pores through a multistep mechanism and provide a new paradigm for understanding the range of different effects of PFN and related membrane attack complex/perforin domain proteins observed in vivo and in vitro.


Subject(s)
Cell Membrane/chemistry , Lipid Bilayers/chemistry , Perforin/chemistry , Cell Membrane/metabolism , Cryoelectron Microscopy , Humans , Lipid Bilayers/metabolism , Perforin/metabolism
9.
J Biol Chem ; 285(29): 22461-72, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20388708

ABSTRACT

The cytoplasm and the nucleus have been identified as activity sites for granzyme B (GrB) following its delivery from cytotoxic lymphocyte granules into target cells. Here we report on the ability of exogenous GrB to insert into and function within a proteinase K-resistant mitochondrial compartment. We identified Hax-1 (HS-1-associated protein X-1), a mitochondrial protein involved in the maintenance of mitochondrial membrane potential, as a GrB substrate within the mitochondrion. GrB cleaves Hax-1 into two major fragments: an N-terminal fragment that localizes to mitochondria and a C-terminal fragment that localizes to the cytosol after being released from GrB-treated mitochondria. The N-terminal Hax-1 fragment major cellular impact is on the regulation of mitochondrial polarization. Overexpression of wild-type Hax-1 or its uncleavable mutant form protects the mitochondria against GrB or valinomycin-mediated depolarization. The N-terminal Hax-1 fragment functions as a dominant negative form of Hax-1, mediating mitochondrial depolarization in a cyclophilin D-dependent manner. Thus, induced expression of the N-terminal Hax-1 fragment results in mitochondrial depolarization and subsequent lysosomal degradation of such altered mitochondria. This study is the first to demonstrate GrB activity within the mitochondrion and to identify Hax-1 cleavage as a novel mechanism for GrB-mediated mitochondrial depolarization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Granzymes/metabolism , Membrane Potential, Mitochondrial , Mitochondria/enzymology , Adaptor Proteins, Signal Transducing/chemistry , Cell Compartmentation/drug effects , Cell Line, Tumor , Peptidyl-Prolyl Isomerase F , Cyclophilins/pharmacology , Gene Knockdown Techniques , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Peptide Fragments/metabolism , Protein Transport/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tetracycline/pharmacology
10.
J Biol Chem ; 285(24): 18918-27, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20395300

ABSTRACT

Granule-associated perforin and granzymes (gzms) are key effector molecules of cytotoxic T lymphocytes (Tc cells) and natural killer cells and play a critical role in the control of intracellular pathogens and cancer. Based on the notion that many gzms, including A, B, C, K, H, and M exhibit cytotoxic activity in vitro, all gzms are believed to serve a similar function in vivo. However, more recent evidence supports the concept that gzms are not unidimensional but, rather, possess non-cytotoxic potential, including stimulation of pro-inflammatory cytokines and anti-viral activities. The present study shows that isolated mouse gzmB cleaves the actin-severing mouse protein, cytoplasmic gelsolin (c-gelsolin) in vitro. However, when delivered to intact target cells by ex vivo immune Tc cells, gzmB mediates c-gelsolin proteolysis via activation of caspases 3/7. The NH(2)-terminal c-gelsolin fragment generated by either gzmB or caspase 3 in vitro constitutively severs actin filaments without destroying the target cells. The observation that gzmB secreted by Tc cells initiates a caspase cascade that disintegrates the actin cytoskeleton in target cells suggests that this intracellular process may contribute to anti-viral host defense.


Subject(s)
Caspase 3/metabolism , Cytoskeleton/metabolism , Gelsolin/chemistry , Granzymes/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Animals , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Fibroblasts/metabolism , Gelsolin/metabolism , Lymphocytic choriomeningitis virus/metabolism , Mice , Microscopy, Fluorescence/methods , Models, Biological , RNA, Messenger/metabolism , Transcription, Genetic
11.
J Cell Biol ; 160(6): 875-85, 2003 Mar 17.
Article in English | MEDLINE | ID: mdl-12629051

ABSTRACT

Granzyme B (GrB), acting similar to an apical caspase, efficiently activates a proteolytic cascade after intracellular delivery by perforin. Studies here were designed to learn whether the physiologic effector, GrB-serglycin, initiates apoptosis primarily through caspase-3 or through BH3-only proteins with subsequent mitochondrial permeabilization and apoptosis. Using four separate cell lines that were either genetically lacking the zymogen or rendered deficient in active caspase-3, we measured apoptotic indices within whole cells (active caspase-3, mitochondrial depolarization [DeltaPsim] and TUNEL). Adhering to these conditions, the following were observed in targets after GrB delivery: (a) procaspase-3-deficient cells fail to display a reduced DeltaPsim and DNA fragmentation; (b) Bax/Bak is required for optimal DeltaPsim reduction, caspase-3 activation, and DNA fragmentation, whereas BID cleavage is undetected by immunoblot; (c) Bcl-2 inhibits GrB-mediated apoptosis (reduced DeltaPsim and TUNEL reactivity) by blocking oligomerization of caspase-3; and (d) in procaspase-3-deficient cells a mitochondrial-independent pathway was identified which involved procaspase-7 activation, PARP cleavage, and nuclear condensation. The data therefore support the existence of a fully implemented apoptotic pathway initiated by GrB, propagated by caspase-3, and perpetuated by a mitochondrial amplification loop but also emphasize the presence of an ancillary caspase-dependent, mitochondria-independent pathway.


Subject(s)
Apoptosis/physiology , Caspases/deficiency , Enzyme Precursors/deficiency , Mitochondria/enzymology , Serine Endopeptidases/deficiency , T-Lymphocytes, Cytotoxic/enzymology , Animals , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3 , Caspase 7 , Caspases/genetics , Caspases/metabolism , DNA Fragmentation/physiology , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Fibroblasts , Granzymes , Humans , Jurkat Cells , Membrane Potentials/physiology , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Serine Endopeptidases/genetics , Signal Transduction/physiology , T-Lymphocytes, Cytotoxic/cytology , bcl-2-Associated X Protein
12.
J Cell Biol ; 160(2): 223-33, 2003 Jan 20.
Article in English | MEDLINE | ID: mdl-12538642

ABSTRACT

The 280-kD cation-independent mannose-6-phosphate receptor (MPR) has been shown to play a role in endocytic uptake of granzyme B, since target cells overexpressing MPR have an increased sensitivity to granzyme B-mediated apoptosis. On this basis, it has been proposed that cells lacking MPR are poor targets for cytotoxic lymphocytes that mediate allograft rejection or tumor immune surveillance. In the present study, we report that the uptake of granzyme B into target cells is independent of MPR. We used HeLa cells overexpressing a dominant-negative mutated (K44A) form of dynamin and mouse fibroblasts overexpressing or lacking MPR to show that the MPR/clathrin/dynamin pathway is not required for granzyme B uptake. Consistent with this observation, cells lacking the MPR/clathrin pathway remained sensitive to granzyme B. Exposure of K44A-dynamin-overexpressing and wild-type HeLa cells to granzyme B with sublytic perforin resulted in similar apoptosis in the two cell populations, both in short and long term assays. Granzyme B uptake into MPR-overexpressing L cells was more rapid than into MPR-null L cells, but the receptor-deficient cells took up granzyme B through fluid phase micropinocytosis and remained sensitive to it. Contrary to previous findings, we also demonstrated that mouse tumor allografts that lack MPR expression were rejected as rapidly as tumors that overexpress MPR. Entry of granzyme B into target cells and its intracellular trafficking to induce target cell death in the presence of perforin are therefore not critically dependent on MPR or clathrin/dynamin-dependent endocytosis.


Subject(s)
Apoptosis/immunology , Cell Membrane/immunology , Endocytosis/immunology , Killer Cells, Natural/enzymology , Receptor, IGF Type 2/deficiency , Serine Endopeptidases/immunology , T-Lymphocytes, Cytotoxic/enzymology , Animals , Apoptosis/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Clathrin/drug effects , Clathrin/genetics , Clathrin/metabolism , Dynamins/drug effects , Dynamins/genetics , Dynamins/metabolism , Endocytosis/drug effects , Female , Graft Rejection/genetics , Graft Rejection/immunology , Granzymes , HeLa Cells , Humans , Killer Cells, Natural/immunology , Male , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neoplasms/immunology , Neoplasms/metabolism , Perforin , Pore Forming Cytotoxic Proteins , Receptor, IGF Type 2/drug effects , Receptor, IGF Type 2/genetics , Serine Endopeptidases/deficiency , Serine Endopeptidases/pharmacology , T-Lymphocytes, Cytotoxic/immunology
13.
Biochem Biophys Res Commun ; 371(3): 391-4, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18439904

ABSTRACT

Granzyme B (GrB), a component of the cytotoxic cell granule secretion pathway, is designed to kill infected and transformed cells after intracellular delivery by the pore forming protein, perforin. The mechanism of the delivery remains speculative. In this study we tested the hypothesis that GrB possesses capacity to bind and disrupt lipid membranes. Here in comparison to previous studies that show GrB interacts with carbohydrate moieties, the protease does not bind membrane phospholipids nor has intrinsic membranolytic properties. To study the transmembrane movement of GrB, we developed a model membrane system consisting of a high-molecular weight GrB substrate encapsulated in unilamellar vesicles. Intra-vesicle proteolysis clearly requires concentrations of lytic agents (streptolysin O, perforin or Triton X-100) that disrupt unilamellar membranes.


Subject(s)
Granzymes/chemistry , Lipids/chemistry , Models, Chemical , Unilamellar Liposomes/chemistry , Bacterial Proteins/chemistry , Octoxynol/chemistry , Perforin/chemistry , Streptolysins/chemistry
14.
Curr Opin Immunol ; 15(5): 528-32, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14499261

ABSTRACT

The molecular details of cytotoxic granule-mediated apoptosis have been gleaned from the study of the effects of isolated granzymes and perforin on target cells. Recent evidence indicates that the physiological apoptosis-inducing form is a multi-component macro-complex consisting of cationic granule proteins non-covalently linked to the chondroitin-sulfate proteoglycan, serglycin.


Subject(s)
Apoptosis , Cytoplasmic Granules/metabolism , Animals , Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfate Proteoglycans/physiology , Cytoplasmic Granules/physiology , Cytotoxicity, Immunologic , Exocytosis , Granzymes , Humans , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Models, Immunological , Perforin , Pore Forming Cytotoxic Proteins , Proteoglycans/physiology , Serine Endopeptidases/physiology , Signal Transduction , T-Lymphocytes, Cytotoxic/metabolism , Vesicular Transport Proteins
15.
Arthritis Rheumatol ; 69(2): 320-334, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27598995

ABSTRACT

OBJECTIVE: Granzyme A (GzmA) levels are elevated in the plasma and synovium of patients with rheumatoid arthritis (RA), suggesting involvement of this protease in the pathogenesis of the disease. GzmA contributes to sepsis by regulating the production of proinflammatory cytokines. The purpose of this study was to evaluate the contribution of GzmA to the pathogenesis of RA in vivo and to examine the possibility that GzmA acting via tumor necrosis factor (TNF) stimulates osteoclastogenesis. METHODS: Inflammatory arthritis induced by type II collagen was evaluated in wild-type, GzmA-deficient, and perforin-deficient mice. The osteoclastogenic potential of GzmA was examined in vitro using bone marrow cells and colony-forming unit-granulocyte-macrophage (CFU-GM) cells and in vivo using GzmA-deficient mice. RESULTS: Gene deletion of GzmA attenuated collagen-induced arthritis, including serum levels of proinflammatory cytokines, joint damage, and bone erosion in affected mice, suggesting that osteoclast activity is reduced in the absence of GzmA. Accordingly, GzmA-treated bone marrow cells produced multinucleated cells that fulfilled the criteria for mature osteoclasts: tartrate-resistant acid phosphatase (TRAP) activity, ß integrin expression, calcitonin receptor expression, and resorptive activity on dentin slices. GzmA appeared to act without accessory cells, and its activity was not affected by osteoprotegerin, suggesting a minor contribution of RANKL. It also induced the expression and secretion of TNF. Neutralization of TNF or stimulation of CFU-GM cells from TNF-/- mice prevented GzmA-induced osteoclastogenesis. GzmA-deficient mice had reduced osteoclastogenesis in vivo (fewer calcitonin receptor-positive multinucleated cells and fewer transcripts for cathepsin K, matrix metalloproteinase 9, and TRAP in joints) and reduced serum levels of C-terminal telopeptide of type I collagen. CONCLUSION: GzmA contributes to the joint destruction of RA partly by promoting osteoclast differentiation.


Subject(s)
Arthritis, Experimental/enzymology , Arthritis, Experimental/etiology , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/etiology , Granzymes/physiology , Osteogenesis/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Female , Mice , Mice, Inbred C57BL
17.
Cell Death Discov ; 2: 16084, 2016.
Article in English | MEDLINE | ID: mdl-28028441

ABSTRACT

Granzymes are serine proteases that, upon release from cytotoxic cells, induce apoptosis in tumor cells and virally infected cells. In addition, a role of granzymes in inflammation is emerging. Recently, we have demonstrated that extracellular granzyme K (GrK) potentiates lipopolysaccharide (LPS)-induced cytokine response from monocytes. GrK interacts with LPS, disaggregates LPS micelles, and stimulates LPS-CD14 binding and Toll-like receptor signaling. Here we show that human GrA also potentiates cytokine responses in human monocytes initiated by LPS or Gram-negative bacteria. Similar to GrK, this effect is independent of GrA catalytic activity. Unlike GrK, however, GrA does not bind to LPS, has little influence on LPS micelle disaggregation, and does not augment LPS-CD14 complex formation. We conclude that GrA and GrK differentially modulate LPS-Toll-like receptor signaling in monocytes, suggesting functional redundancy among cytotoxic lymphocyte proteases in the anti-bacterial innate immune response.

18.
J Immunol Methods ; 299(1-2): 117-27, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15914196

ABSTRACT

How perforin (PFN) delivers the granzymes during cytotoxic granule mediated apoptosis remains a mystery. A major obstacle has been the inability to visualize PFN in either monomeric or polymeric form after interaction with the target cell surface. An antibody based technique is described which detects cell surface PFN on intact cells by flow cytometry. The methodology requires the presence of calcium (Ca2+) at a concentration which supports binding but not polymerization of PFN. Functionality was ensured by showing the cell surface PFN was able to deliver GrB causing caspase-3 activation and mitochondrial depolarization. The technique demonstrates a role for heparan sulfate proteoglycans in PFN binding. Further, the variable sensitivity of effector versus target cell lines to the permeabilizing effects of PFN could not be attributed to differential binding of PFN.


Subject(s)
Flow Cytometry/methods , Membrane Glycoproteins/analysis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Biological Assay , Calcium/pharmacology , Cell Membrane/chemistry , Cell Membrane/drug effects , Heparan Sulfate Proteoglycans/pharmacology , Humans , Hydrogen-Ion Concentration , Jurkat Cells , Membrane Glycoproteins/immunology , Membrane Glycoproteins/pharmacology , Perforin , Permeability/drug effects , Pore Forming Cytotoxic Proteins
19.
Clin Cancer Res ; 10(21): 7402-12, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15534117

ABSTRACT

PURPOSE: Because WT1 is a universal tumor antigen, we examined the sensitivity of myeloma cells to WT1-specific cytotoxic T lymphocyte (CTL)-mediated cytotoxicity. EXPERIMENTAL DESIGN: WT1 expression in hematologic malignant cells was examined by quantitative reverse transcription-polymerase chain reaction. The cytotoxicity of a WT1-specific CTL clone against hematologic malignant cells, including myeloma cells, was examined by standard chromium-51 release assays. The extent of membrane damage induced by purified perforin was examined. Induction of WT1-specific CTLs from the patients with multiple myeloma (MM) was attempted, and we examined their function against myeloma cells. RESULTS: The expression levels of WT1 mRNA in myeloma and lymphoma cells were significantly lower than that in acute leukemia cells. Although the WT1 expression levels in myeloma and lymphoma cells were almost same, only myeloma cells were lysed efficiently by WT1-specific CTLs in a HLA-restricted manner. The amounts of interferon-gamma produced by WT1-specific CTLs in response to stimulation with myeloma cells and with lymphoma cells were almost the same, suggesting that WT1 protein is processed and expressed in the context of HLA class I molecules similarly on both myeloma and lymphoma cells. The extent of membrane damage induced by purified perforin appeared to be significantly higher in myeloma cells than in lymphoma cells. WT1-specific CTLs appeared to be present in patients with MM. CONCLUSIONS: The present study has shown that susceptibility of membranes to perforin is an important factor determining the sensitivity of target cells to CTL-mediated cytotoxicity and that WT1 is an ideal target antigen for cellular immunotherapy of MM.


Subject(s)
Cytoplasmic Granules/metabolism , Multiple Myeloma/metabolism , T-Lymphocytes, Cytotoxic/metabolism , WT1 Proteins/biosynthesis , Antigens, Neoplasm/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Calcium/metabolism , Cell Line , Cell Line, Tumor , Chromium/metabolism , Cytoplasmic Granules/physiology , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Enzyme-Linked Immunosorbent Assay , Exocytosis , Flow Cytometry , Genes, MHC Class I , Humans , Immunotherapy , Interferon-gamma/metabolism , Lymphoma/metabolism , Macrolides/pharmacology , Membrane Glycoproteins/metabolism , Peptides/chemistry , Perforin , Pore Forming Cytotoxic Proteins , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
20.
Clin Cancer Res ; 10(6): 1894-900, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15041704

ABSTRACT

PURPOSE AND EXPERIMENTAL DESIGN: We have previously observed that glucose deprivation enhances tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptotic death as well as caspase activation (caspase-3, -9, and -8) in human prostate adenocarcinoma DU-145 cells. In this study, we used caspase-3-deficient MCF-7 breast cancer cells to examine the possible role of caspase-3 in glucose deprivation-enhanced TRAIL cytotoxicity. RESULTS: Combined glucose deprivation and 200 ng/ml TRAIL treatment markedly induced cytotoxicity in caspase-3 cDNA transfected cells (MCF-7/casp-3) but not in control vector transfected cells (MCF-7/vector). We also observed that the level of Akt, an antiapoptotic protein, was reduced by treatment with TRAIL in MCF-7/casp-3 cells but not in MCF-7/vector cells. The reduction of Akt by TRAIL was promoted in the absence of glucose in MCF-7/casp-3 cells. However, pretreatment with 20 micro M Z-LEHD-FMK, a caspase-9 inhibitor, protected MCF-7/casp-3 cells from the combinatorial treatment of TRAIL and glucose deprivation-induced cytotoxicity. This compound also prevented the reduction of Akt level during the combinatorial treatment. Moreover, this Akt reduction was not inhibited by treatment with MG-132, a proteosome inhibitor. Data from site-directed mutagenesis show that Akt was cleaved at amino acid 108, but not 119, during treatment with TRAIL and glucose deprivation. CONCLUSIONS: Our results suggest that caspase-3 is involved in the reduction of Akt level, and its involvement is mediated through caspase-9 activation. The reduction of Akt level is also due to cleavage of Akt rather than degradation of Akt.


Subject(s)
Caspases/metabolism , Glucose/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Breast Neoplasms , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/deficiency , Cell Line, Tumor , Cell Survival , Female , Humans , Male , Mutagenesis, Site-Directed/genetics , Peptide Fragments/pharmacology , Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL