Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1173519, 2023.
Article in English | MEDLINE | ID: mdl-37266429

ABSTRACT

The discovery of gasdermin D (GSDMD) as the terminal executioner of pyroptosis provided a large piece of the cell death puzzle, whilst simultaneously and firmly putting the gasdermin family into the limelight. In its purest form, GSDMD provides a connection between the innate alarm systems to an explosive, inflammatory form of cell death to jolt the local environment into immunological action. However, the gasdermin field has moved rapidly and significantly since the original seminal work and novel functions and mechanisms have been recently uncovered, particularly in response to infection. Gasdermins regulate and are regulated by mechanisms such as autophagy, metabolism and NETosis in fighting pathogen and protecting host. Importantly, activators and interactors of the other gasdermins, not just GSDMD, have been recently elucidated and have opened new avenues for gasdermin-based discovery. Key to this is the development of potent and specific tool molecules, so far a challenge for the field. Here we will cover some of these recently discovered areas in relation to bacterial infection before providing an overview of the pharmacological landscape and the challenges associated with targeting gasdermins.


Subject(s)
Bacteriology , Gasdermins , Intracellular Signaling Peptides and Proteins , Pyroptosis , Cell Death
2.
Eur Respir J ; 61(4)2023 04.
Article in English | MEDLINE | ID: mdl-36549711

ABSTRACT

BACKGROUND: Receptor-interacting protein kinase 1 (RIPK1) is a key mediator of regulated cell death (including apoptosis and necroptosis) and inflammation, both drivers of COPD pathogenesis. We aimed to define the contribution of RIPK1 kinase-dependent cell death and inflammation in the pathogenesis of COPD. METHODS: We assessed RIPK1 expression in single-cell RNA sequencing (RNA-seq) data from human and mouse lungs, and validated RIPK1 levels in lung tissue of COPD patients via immunohistochemistry. Next, we assessed the consequences of genetic and pharmacological inhibition of RIPK1 kinase activity in experimental COPD, using Ripk1 S25D/S25D kinase-deficient mice and the RIPK1 kinase inhibitor GSK'547. RESULTS: RIPK1 expression increased in alveolar type 1 (AT1), AT2, ciliated and neuroendocrine cells in human COPD. RIPK1 protein levels were significantly increased in airway epithelium of COPD patients compared with never-smokers and smokers without airflow limitation. In mice, exposure to cigarette smoke (CS) increased Ripk1 expression similarly in AT2 cells, and further in alveolar macrophages and T-cells. Genetic and/or pharmacological inhibition of RIPK1 kinase activity significantly attenuated airway inflammation upon acute and subacute CS exposure, as well as airway remodelling, emphysema, and apoptotic and necroptotic cell death upon chronic CS exposure. Similarly, pharmacological RIPK1 kinase inhibition significantly attenuated elastase-induced emphysema and lung function decline. Finally, RNA-seq on lung tissue of CS-exposed mice revealed downregulation of cell death and inflammatory pathways upon pharmacological RIPK1 kinase inhibition. CONCLUSIONS: RIPK1 kinase inhibition is protective in experimental models of COPD and may represent a novel promising therapeutic approach.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Mice , Animals , Lung , Cell Death , Inflammation/metabolism , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
3.
Nat Commun ; 12(1): 3364, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099649

ABSTRACT

Necroptosis is a lytic, inflammatory form of cell death that not only contributes to pathogen clearance but can also lead to disease pathogenesis. Necroptosis is triggered by RIPK3-mediated phosphorylation of MLKL, which is thought to initiate MLKL oligomerisation, membrane translocation and membrane rupture, although the precise mechanism is incompletely understood. Here, we show that K63-linked ubiquitin chains are attached to MLKL during necroptosis and that ubiquitylation of MLKL at K219 significantly contributes to the cytotoxic potential of phosphorylated MLKL. The K219R MLKL mutation protects animals from necroptosis-induced skin damage and renders cells resistant to pathogen-induced necroptosis. Mechanistically, we show that ubiquitylation of MLKL at K219 is required for higher-order assembly of MLKL at membranes, facilitating its rupture and necroptosis. We demonstrate that K219 ubiquitylation licenses MLKL activity to induce lytic cell death, suggesting that necroptotic clearance of pathogens as well as MLKL-dependent pathologies are influenced by the ubiquitin-signalling system.


Subject(s)
Herpesviridae Infections/metabolism , Lysine/metabolism , Protein Kinases/metabolism , Skin/metabolism , Animals , Cell Line , Cells, Cultured , HEK293 Cells , HT29 Cells , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Humans , Lysine/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/physiology , NIH 3T3 Cells , Necroptosis/genetics , Necrosis , Protein Kinases/genetics , Skin/pathology , Ubiquitination
6.
Commun Biol ; 3(1): 140, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32198438

ABSTRACT

Proteolysis-Targeting Chimeras (PROTACs) are heterobifunctional small-molecules that can promote the rapid and selective proteasome-mediated degradation of intracellular proteins through the recruitment of E3 ligase complexes to non-native protein substrates. The catalytic mechanism of action of PROTACs represents an exciting new modality in drug discovery that offers several potential advantages over traditional small-molecule inhibitors, including the potential to deliver pharmacodynamic (PD) efficacy which extends beyond the detectable pharmacokinetic (PK) presence of the PROTAC, driven by the synthesis rate of the protein. Herein we report the identification and development of PROTACs that selectively degrade Receptor-Interacting Serine/Threonine Protein Kinase 2 (RIPK2) and demonstrate in vivo degradation of endogenous RIPK2 in rats at low doses and extended PD that persists in the absence of detectable compound. This disconnect between PK and PD, when coupled with low nanomolar potency, offers the potential for low human doses and infrequent dosing regimens with PROTAC medicines.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drug Design , Inflammation/prevention & control , Leukocytes, Mononuclear/drug effects , Proteasome Endopeptidase Complex/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/enzymology , Crohn Disease/drug therapy , Crohn Disease/enzymology , Cytokines/metabolism , Dose-Response Relationship, Drug , Enzyme Stability , Female , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation Mediators/metabolism , Injections, Intravenous , Leukocytes, Mononuclear/enzymology , Male , Proteolysis , Rats, Sprague-Dawley , Rats, Wistar , THP-1 Cells , Tissue Culture Techniques , Ubiquitination
7.
J Med Chem ; 62(14): 6482-6494, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31265286

ABSTRACT

RIP2 kinase has been identified as a key signal transduction partner in the NOD2 pathway contributing to a variety of human pathologies, including immune-mediated inflammatory diseases. Small-molecule inhibitors of RIP2 kinase or its signaling partners on the NOD2 pathway that are suitable for advancement into the clinic have yet to be described. Herein, we report our discovery and profile of the prodrug clinical compound, inhibitor 3, currently in phase 1 clinical studies. Compound 3 potently binds to RIP2 kinase with good kinase specificity and has excellent activity in blocking many proinflammatory cytokine responses in vivo and in human IBD explant samples. The highly favorable physicochemical and ADMET properties of 3 combined with high potency led to a predicted low oral dose in humans.


Subject(s)
Benzothiazoles/pharmacology , Phosphates/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Animals , Benzothiazoles/chemistry , Benzothiazoles/pharmacokinetics , Benzothiazoles/therapeutic use , Colitis/drug therapy , Dogs , Drug Discovery , Humans , Male , Mice , Molecular Docking Simulation , Phosphates/chemistry , Phosphates/pharmacokinetics , Phosphates/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Swine , Swine, Miniature
8.
J Med Chem ; 62(10): 5096-5110, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31013427

ABSTRACT

RIP1 kinase regulates necroptosis and inflammation and may play an important role in contributing to a variety of human pathologies, including inflammatory and neurological diseases. Currently, RIP1 kinase inhibitors have advanced into early clinical trials for evaluation in inflammatory diseases such as psoriasis, rheumatoid arthritis, and ulcerative colitis and neurological diseases such as amyotrophic lateral sclerosis and Alzheimer's disease. In this paper, we report on the design of potent and highly selective dihydropyrazole (DHP) RIP1 kinase inhibitors starting from a high-throughput screen and the lead-optimization of this series from a lead with minimal rat oral exposure to the identification of dihydropyrazole 77 with good pharmacokinetic profiles in multiple species. Additionally, we identified a potent murine RIP1 kinase inhibitor 76 as a valuable in vivo tool molecule suitable for evaluating the role of RIP1 kinase in chronic models of disease. DHP 76 showed efficacy in mouse models of both multiple sclerosis and human retinitis pigmentosa.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Nuclear Pore Complex Proteins/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , Animals , Biological Availability , Cell Line , Chronic Disease , Drug Design , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Enzyme Inhibitors/pharmacokinetics , Haplorhini , High-Throughput Screening Assays , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Multiple Sclerosis/drug therapy , Pyrazoles/pharmacokinetics , Rats , Retinitis Pigmentosa/drug therapy , Structure-Activity Relationship
9.
Cancer Cell ; 34(5): 757-774.e7, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30423296

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is characterized by immune tolerance and immunotherapeutic resistance. We discovered upregulation of receptor-interacting serine/threonine protein kinase 1 (RIP1) in tumor-associated macrophages (TAMs) in PDA. To study its role in oncogenic progression, we developed a selective small-molecule RIP1 inhibitor with high in vivo exposure. Targeting RIP1 reprogrammed TAMs toward an MHCIIhiTNFα+IFNγ+ immunogenic phenotype in a STAT1-dependent manner. RIP1 inhibition in TAMs resulted in cytotoxic T cell activation and T helper cell differentiation toward a mixed Th1/Th17 phenotype, leading to tumor immunity in mice and in organotypic models of human PDA. Targeting RIP1 synergized with PD1-and inducible co-stimulator-based immunotherapies. Tumor-promoting effects of RIP1 were independent of its co-association with RIP3. Collectively, our work describes RIP1 as a checkpoint kinase governing tumor immunity.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Immune Tolerance/immunology , Macrophages/immunology , Pancreatic Neoplasms/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , Immune Tolerance/genetics , L Cells , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , STAT1 Transcription Factor/metabolism , Th1 Cells/cytology , Th17 Cells/cytology
10.
Methods Mol Biol ; 1857: 109-124, 2018.
Article in English | MEDLINE | ID: mdl-30136235

ABSTRACT

RIP1 kinase plays a key role in regulating signaling pathways downstream of a number of innate immune receptors such as TNFRI and TLRs. The discovery of Necrostatin-1 (Nec-1) as a small-molecule inhibitor of RIP1 kinase has been very instrumental in defining the necroptotic and other signalling pathways regulated by RIP1, but certain characteristics of Nec-1 limits its utility in experimental systems. Next generation RIP1 kinase inhibitors have been identified and the use of these tool inhibitors along with Nec-1 has revealed that RIP1 is emerging as a key driver of inflammation and tissue injury in the pathogenesis of various diseases. Further studying the role of RIP1 to carefully unravel the complex biology requires the selection of the correct tool small-molecule inhibitors. In addition, it is important to consider the proper application of current tool inhibitors and understand the current limitiations. Here we will discuss key parameters that need to be considered when selecting and applying tool inhibitors to novel biological assays and systems. General protocols to explore the in vitro and in vivo potency, cellular selectivity, and pharmacokinetic properties of current small-molecule inhibitors of RIP1 kinase are provided.


Subject(s)
Apoptosis , Fibrosarcoma/pathology , GTPase-Activating Proteins/antagonists & inhibitors , Macrophages/pathology , Necrosis , Small Molecule Libraries/pharmacology , Animals , Fibrosarcoma/drug therapy , Fibrosarcoma/enzymology , Macrophages/drug effects , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Signal Transduction , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
11.
J Med Chem ; 59(10): 4867-80, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27109867

ABSTRACT

RIP2 kinase is a central component of the innate immune system and enables downstream signaling following activation of the pattern recognition receptors NOD1 and NOD2, leading to the production of inflammatory cytokines. Recently, several inhibitors of RIP2 kinase have been disclosed that have contributed to the fundamental understanding of the role of RIP2 in this pathway. However, because they lack either broad kinase selectivity or strong affinity for RIP2, these tools have only limited utility to assess the role of RIP2 in complex environments. We present, herein, the discovery and pharmacological characterization of GSK583, a next-generation RIP2 inhibitor possessing exquisite selectivity and potency. Having demonstrated the pharmacological precision of this tool compound, we report its use in elucidating the role of RIP2 kinase in a variety of in vitro, in vivo, and ex vivo experiments, further clarifying our understanding of the role of RIP2 in NOD1 and NOD2 mediated disease pathogenesis.


Subject(s)
Aminoquinolines/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Sulfones/pharmacology , Aminoquinolines/blood , Aminoquinolines/chemistry , Animals , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Structure-Activity Relationship , Sulfones/blood , Sulfones/chemistry
12.
PLoS One ; 10(5): e0127083, 2015.
Article in English | MEDLINE | ID: mdl-25965667

ABSTRACT

CARMA-BCL10-MALT1 signalosomes play important roles in antigen receptor signaling and other pathways. Previous studies have suggested that as part of this complex, MALT1 functions as both a scaffolding protein to activate NF-κB through recruitment of ubiquitin ligases, and as a protease to cleave and inactivate downstream inhibitory signaling proteins. However, our understanding of the relative importance of these two distinct MALT1 activities has been hampered by a lack of selective MALT1 protease inhibitors with suitable pharmacologic properties. To fully investigate the role of MALT1 protease activity, we generated mice homozygous for a protease-dead mutation in MALT1. We found that some, but not all, MALT1 functions in immune cells were dependent upon its protease activity. Protease-dead mice had defects in the generation of splenic marginal zone and peritoneal B1 B cells. CD4+ and CD8+ T cells displayed decreased T cell receptor-stimulated proliferation and IL-2 production while B cell receptor-stimulated proliferation was partially dependent on protease activity. In dendritic cells, stimulation of cytokine production through the Dectin-1, Dectin-2, and Mincle C-type lectin receptors was also found to be partially dependent upon protease activity. In vivo, protease-dead mice had reduced basal immunoglobulin levels, and showed defective responses to immunization with T-dependent and T-independent antigens. Surprisingly, despite these decreased responses, MALT1 protease-dead mice, but not MALT1 null mice, developed mixed inflammatory cell infiltrates in multiple organs, suggesting MALT1 protease activity plays a role in immune homeostasis. These findings highlight the importance of MALT1 protease activity in multiple immune cell types, and in integrating immune responses in vivo.


Subject(s)
Caspases/genetics , Caspases/metabolism , Immunity, Innate , Lymphocyte Activation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Animals , B-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Knock-In Techniques , Inflammation/genetics , Inflammation/immunology , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Mutation , Spleen/immunology , T-Lymphocytes/immunology
13.
J Immunol ; 191(4): 1536-46, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23851689

ABSTRACT

Although the pathways that permit IL-2 production and the full activation of T cells upon Ag encounter are fairly well defined, the negative regulatory circuits that limit these pathways are poorly understood. In this study, we show that the E3 ubiquitin ligase adaptor Ndfip1 directs one such negative regulatory circuit. T cells lacking Ndfip1 produce IL-2, upregulate IL-2Rα, and proliferate, in the absence of CD28 costimulation. Furthermore, T cells in mice lacking both Ndfip1 and CD28 become activated, produce IL-4, and drive inflammation at barrier surfaces. Ndfip1 constrains T cell activation by limiting the duration of IL-2 mRNA expression after TCR stimulation. Ndfip1 and IL-2 have a similar expression pattern, and, following TCR stimulation, expression of both Ndfip1 and IL-2 requires the activity of NFAT and Erk. Taken together, these data support a negative regulatory circuit in which factors that induce IL-2 expression downstream of TCR engagement also induce the expression of Ndfip1 to limit the extent of IL-2 production and, thus, dampen T cell activation.


Subject(s)
CD28 Antigens/immunology , Carrier Proteins/physiology , Gene Expression Regulation/immunology , Interleukin-2/biosynthesis , Lymphocyte Activation/immunology , Membrane Proteins/physiology , Animals , Antigen Presentation , Antigens/immunology , CD28 Antigens/biosynthesis , CD28 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Costimulatory and Inhibitory T-Cell Receptors/immunology , Cyclosporine/pharmacology , Feedback, Physiological , Hyaluronan Receptors/analysis , Intercellular Signaling Peptides and Proteins , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Lymphocyte Activation/drug effects , Lymphoid Tissue/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Membrane Proteins/deficiency , Mice , Mice, Knockout , Mice, Transgenic , NFATC Transcription Factors/immunology , Ovalbumin/immunology , Protein Kinase Inhibitors/pharmacology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/physiology
14.
J Immunol ; 188(8): 4023-31, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22403444

ABSTRACT

Ndfip1 is an adaptor for the E3 ubiquitin ligase Itch. Both Ndfip1- and Itch-deficient T cells are biased toward Th2 cytokine production. In this study, we demonstrate that lungs from Ndfip1(-/-) mice showed increased numbers of neutrophils and Th17 cells. This was not because Ndfip1(-/-) T cells are biased toward Th17 differentiation. In fact, fewer Ndfip1(-/-) T cells differentiated into Th17 cells in vitro due to high IL-4 production. Rather, Th17 differentiation was increased in Ndfip1(-/-) mice due to increased numbers of IL-6-producing eosinophils. IL-6 levels in mice that lacked both Ndfip1 and IL-4 were similar to wild-type controls, and these mice had fewer Th17 cells in their lungs. These results indicate that Th2 inflammation, such as that observed in Ndfip1(-/-) mice, can increase Th17 differentiation by recruiting IL-6-producing eosinophils into secondary lymphoid organs and tissues. This may explain why Th17 cells develop within an ongoing Th2 inflammatory response.


Subject(s)
Carrier Proteins/immunology , Lung/immunology , Membrane Proteins/immunology , Th17 Cells/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Carrier Proteins/genetics , Cell Differentiation , Cell Movement/immunology , Eosinophils/immunology , Eosinophils/pathology , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins , Interleukin-4/biosynthesis , Interleukin-4/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lung/pathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Signal Transduction , Th1-Th2 Balance , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Ubiquitin-Protein Ligases/genetics
15.
Nat Immunol ; 13(1): 77-85, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22080920

ABSTRACT

Mice deficient in the adaptor Ndfip1 develop inflammation at sites of environmental antigen exposure. We show here that such mice had fewer inducible regulatory T cells (iT(reg) cells). In vitro, Ndfip1-deficient T cells expressed normal amounts of the transcription factor Foxp3 during the first 48 h of iT(reg) cell differentiation; however, this expression was not sustained. Abortive Foxp3 expression was caused by production of interleukin 4 (IL-4) by Ndfip1(-/-) cells. We found that Ndfip1 expression was transiently upregulated during iT(reg) cell differentiation in a manner dependent on transforming growth factor-ß (TGF-ß). Once expressed, Ndfip1 promoted degradation of the transcription factor JunB mediated by the E3 ubiquitin ligase Itch, thus preventing IL-4 production. On the basis of our data, we propose that TGF-ß signaling induces Ndfip1 expression to silence IL-4 production, thus permitting iT(reg) cell differentiation.


Subject(s)
Carrier Proteins/genetics , Cell Differentiation , Interleukin-4/biosynthesis , Membrane Proteins/genetics , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins , Interleukin-4/genetics , Interleukin-4/pharmacology , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
16.
Immunity ; 31(4): 632-42, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19833088

ABSTRACT

Cytolytic granules mediate killing of virus-infected cells by cytotoxic T lymphocytes. We show here that the granules can take long or short paths to the secretory domain. Both paths utilized the same intracellular molecular events, which have different spatial and temporal arrangements and are regulated by the kinetics of Ca(2+)-mediated signaling. Rapid signaling caused swift granule concentration near the microtubule-organizing center (MTOC) and subsequent delivery by the polarized MTOC directly to the secretory domain-the shortest path. Indolent signaling led to late recruitment of granules that moved along microtubules to the periphery of the synapse and then moved tangentially to fuse at the outer edge of the secretory domain-a longer path. The short pathway is associated with faster granule release and more efficient killing than the long pathway. Thus, the kinetics of early signaling regulates the quality of the T cell cytolytic response.


Subject(s)
Cytoplasmic Granules/immunology , Immunological Synapses/immunology , Microtubule-Organizing Center/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Biological Transport/drug effects , Biological Transport/immunology , Calcium/immunology , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/immunology , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line , Cell Polarity/drug effects , Cell Polarity/immunology , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Humans , Immunological Synapses/drug effects , Immunological Synapses/metabolism , Ionomycin/pharmacology , Ionophores/pharmacology , Microtubule-Organizing Center/drug effects , Microtubule-Organizing Center/metabolism , Microtubules/drug effects , Microtubules/immunology , Microtubules/metabolism , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism
17.
J Immunol ; 181(7): 4815-24, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802085

ABSTRACT

Destruction of virus-infected cells by CTL is an extremely sensitive and efficient process. Our previous data suggest that LFA-1-ICAM-1 interactions in the peripheral supramolecular activation cluster (pSMAC) of the immunological synapse mediate formation of a tight adhesion junction that might contribute to the sensitivity of target cell lysis by CTL. Herein, we compared more (CD8(+)) and less (CD4(+)) effective CTL to understand the molecular events that promote efficient target cell lysis. We found that abrogation of the pSMAC formation significantly impaired the ability of CD8(+) but not CD4(+) CTL to lyse target cells despite having no effect of the amount of released granules by both CD8(+) and CD4(+) CTL. Consistent with this, CD4(+) CTL break their synapses more often than do CD8(+) CTL, which leads to the escape of the cytolytic molecules from the interface. CD4(+) CTL treatment with a protein kinase Ctheta inhibitor increases synapse stability and sensitivity of specific target cell lysis. Thus, formation of a stable pSMAC, which is partially controlled by protein kinase Ctheta, functions to confine the released lytic molecules at the synaptic interface and to enhance the effectiveness of target cell lysis.


Subject(s)
Cytotoxicity Tests, Immunologic , Immunological Synapses/enzymology , Immunological Synapses/immunology , Isoenzymes/physiology , Protein Kinase C/physiology , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Adhesion/immunology , Cell Aggregation/immunology , Cell Communication/immunology , Cell Line, Transformed , Clone Cells , Cytotoxicity Tests, Immunologic/methods , Enzyme Stability/immunology , HIV Antigens/immunology , HIV Antigens/metabolism , Humans , Immunological Synapses/metabolism , Influenza A virus/immunology , Lymphocyte Activation/immunology , Molecular Sequence Data , Protein Kinase C-theta , T-Lymphocytes, Cytotoxic/virology , Viral Matrix Proteins/immunology , Viral Matrix Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...