Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 197(4): 1353-67, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27385778

ABSTRACT

Canonical inflammasome activation induces a caspase-1/gasdermin D (Gsdmd)-dependent lytic cell death called pyroptosis that promotes antimicrobial host defense but may contribute to sepsis. The nature of the caspase-1-dependent change in plasma membrane (PM) permeability during pyroptotic progression remains incompletely defined. We assayed propidium(2+) (Pro(2+)) influx kinetics during NLRP3 or Pyrin inflammasome activation in murine bone marrow-derived macrophages (BMDMs) as an indicator of this PM permeabilization. BMDMs were characterized by rapid Pro(2+) influx after initiation of NLRP3 or Pyrin inflammasomes by nigericin (NG) or Clostridium difficile toxin B (TcdB), respectively. No Pro(2+) uptake in response to NG or TcdB was observed in Casp1(-/-) or Asc(-/-) BMDMs. The cytoprotectant glycine profoundly suppressed NG and TcdB-induced lysis but not Pro(2+) influx. The absence of Gsdmd expression resulted in suppression of NG-stimulated Pro(2+) influx and pyroptotic lysis. Extracellular La(3+) and Gd(3+) rapidly and reversibly blocked the induced Pro(2+) influx and markedly delayed pyroptotic lysis without limiting upstream inflammasome assembly and caspase-1 activation. Thus, caspase-1-driven pyroptosis requires induction of initial prelytic pores in the PM that are dependent on Gsdmd expression. These PM pores also facilitated the efflux of cytosolic ATP and influx of extracellular Ca(2+) Although lanthanides and Gsdmd deletion both suppressed PM pore activity and pyroptotic lysis, robust IL-1ß release was observed in lanthanide-treated BMDMs but not in Gsdmd-deficient cells. This suggests roles for Gsdmd in both passive IL-1ß release secondary to pyroptotic lysis and in nonlytic/nonclassical IL-1ß export.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Pyroptosis/physiology , Animals , Cell Membrane/pathology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Lanthanoid Series Elements/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins
2.
Am J Physiol Cell Physiol ; 311(1): C83-C100, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27170638

ABSTRACT

Nucleotide-binding domain, leucine-rich-repeat-containing family, pyrin domain-containing 3 (NLRP3) is a cytosolic protein that nucleates assembly of inflammasome signaling platforms, which facilitate caspase-1-mediated IL-1ß release and other inflammatory responses in myeloid leukocytes. NLRP3 inflammasomes are assembled in response to multiple pathogen- or environmental stress-induced changes in basic cell physiology, including the destabilization of lysosome integrity and activation of K(+)-permeable channels/transporters in the plasma membrane (PM). However, the quantitative relationships between lysosome membrane permeabilization (LMP), induction of increased PM K(+) permeability, and activation of NLRP3 signaling are incompletely characterized. We used Leu-Leu-O-methyl ester (LLME), a soluble lysosomotropic agent, to quantitatively track the kinetics and extent of LMP in relation to NLRP3 inflammasome signaling responses (ASC oligomerization, caspase-1 activation, IL-1ß release) and PM cation fluxes in murine bone marrow-derived dendritic cells (BMDCs). Treatment of BMDCs with submillimolar (≤1 mM) LLME induced slower and partial increases in LMP that correlated with robust NLRP3 inflammasome activation and K(+) efflux. In contrast, supramillimolar (≥2 mM) LLME elicited extremely rapid and complete collapse of lysosome integrity that was correlated with suppression of inflammasome signaling. Supramillimolar LLME also induced dominant negative effects on inflammasome activation by the canonical NLRP3 agonist nigericin; this inhibition correlated with an increase in NLRP3 ubiquitination. LMP elicited rapid BMDC death by both inflammasome-dependent pyroptosis and inflammasome-independent necrosis. LMP also triggered Ca(2+) influx, which attenuated LLME-stimulated NLRP3 inflammasome signaling but potentiated LLME-induced necrosis. Taken together, these studies reveal a previously unappreciated signaling network that defines the coupling between LMP, changes in PM cation fluxes, cell death, and NLRP3 inflammasome activation.


Subject(s)
Calcium Signaling , Calcium/metabolism , Dendritic Cells/metabolism , Inflammasomes/metabolism , Lysosomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , Calcium Signaling/drug effects , Caspase 1/deficiency , Caspase 1/genetics , Caspases/deficiency , Caspases/genetics , Caspases, Initiator , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/pathology , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Inflammasomes/drug effects , Interleukin-1beta/metabolism , Kinetics , Lysosomes/drug effects , Lysosomes/pathology , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Nigericin/pharmacology , Permeability , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , Ubiquitination
3.
Nat Commun ; 7: 10555, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26877061

ABSTRACT

Although extracellular ATP is abundant at sites of inflammation, its role in activating inflammasome signalling in neutrophils is not well characterized. In the current study, we demonstrate that human and murine neutrophils express functional cell-surface P2X7R, which leads to ATP-induced loss of intracellular K(+), NLRP3 inflammasome activation and IL-1ß secretion. ATP-induced P2X7R activation caused a sustained increase in intracellular [Ca(2+)], which is indicative of P2X7R channel opening. Although there are multiple polymorphic variants of P2X7R, we found that neutrophils from multiple donors express P2X7R, but with differential efficacies in ATP-induced increase in cytosolic [Ca(2+)]. Neutrophils were also the predominant P2X7R-expressing cells during Streptococcus pneumoniae corneal infection, and P2X7R was required for bacterial clearance. Given the ubiquitous presence of neutrophils and extracellular ATP in multiple inflammatory conditions, ATP-induced P2X7R activation and IL-1ß secretion by neutrophils likely has a significant, wide ranging clinical impact.


Subject(s)
Adenosine Triphosphate/immunology , Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/metabolism , Neutrophils/immunology , Receptors, Purinergic P2X7/immunology , Animals , Blotting, Western , Calcium/metabolism , Carrier Proteins/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Eye Infections, Bacterial/immunology , Flow Cytometry , Humans , Keratitis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils/drug effects , Neutrophils/metabolism , Potassium/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Spectrophotometry, Atomic , Streptococcal Infections/immunology
4.
J Immunol ; 194(8): 3937-52, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25762778

ABSTRACT

Perturbation of intracellular ion homeostasis is a major cellular stress signal for activation of NLRP3 inflammasome signaling that results in caspase-1-mediated production of IL-1ß and pyroptosis. However, the relative contributions of decreased cytosolic K(+) concentration versus increased cytosolic Ca(2+) concentration ([Ca(2+)]) remain disputed and incompletely defined. We investigated roles for elevated cytosolic [Ca(2+)] in NLRP3 activation and downstream inflammasome signaling responses in primary murine dendritic cells and macrophages in response to two canonical NLRP3 agonists (ATP and nigericin) that facilitate primary K(+) efflux by mechanistically distinct pathways or the lysosome-destabilizing agonist Leu-Leu-O-methyl ester. The study provides three major findings relevant to this unresolved area of NLRP3 regulation. First, increased cytosolic [Ca(2+)] was neither a necessary nor sufficient signal for the NLRP3 inflammasome cascade during activation by endogenous ATP-gated P2X7 receptor channels, the exogenous bacterial ionophore nigericin, or the lysosomotropic agent Leu-Leu-O-methyl ester. Second, agonists for three Ca(2+)-mobilizing G protein-coupled receptors (formyl peptide receptor, P2Y2 purinergic receptor, and calcium-sensing receptor) expressed in murine dendritic cells were ineffective as activators of rapidly induced NLRP3 signaling when directly compared with the K(+) efflux agonists. Third, the intracellular Ca(2+) buffer, BAPTA, and the channel blocker, 2-aminoethoxydiphenyl borate, widely used reagents for disruption of Ca(2+)-dependent signaling pathways, strongly suppressed nigericin-induced NLRP3 inflammasome signaling via mechanisms dissociated from their canonical or expected effects on Ca(2+) homeostasis. The results indicate that the ability of K(+) efflux agonists to activate NLRP3 inflammasome signaling can be dissociated from changes in cytosolic [Ca(2+)] as a necessary or sufficient signal.


Subject(s)
Calcium Signaling/immunology , Carrier Proteins/immunology , Inflammasomes/immunology , Potassium/immunology , Adenosine Triphosphate/immunology , Animals , Boron Compounds , Calcium Signaling/drug effects , Chelating Agents/pharmacology , Dipeptides/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-1beta/immunology , Ionophores/pharmacology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nigericin/pharmacology , Receptors, Purinergic P2X7/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...