Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Nat Immunol ; 13(4): 325-32, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22430785

ABSTRACT

Inflammasomes are multiprotein complexes that activate caspase-1, which leads to maturation of the proinflammatory cytokines interleukin 1ß (IL-1ß) and IL-18 and the induction of pyroptosis. Members of the Nod-like receptor (NLR) family, including NLRP1, NLRP3 and NLRC4, and the cytosolic receptor AIM2 are critical components of inflammasomes and link microbial and endogenous danger signals to the activation of caspase-1. In response to microbial infection, activation of the inflammasomes contributes to host protection by inducing immune responses that limit microbial invasion, but deregulated activation of inflammasomes is associated with autoinflammatory syndromes and other pathologies. Thus, understanding inflammasome pathways may provide insight into the mechanisms of host defense against microbes and the development of inflammatory disorders.


Subject(s)
Immunity, Innate/immunology , Inflammasomes/immunology , Signal Transduction/immunology , Animals , Humans
2.
Nat Immunol ; 13(5): 449-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22484733

ABSTRACT

Intestinal phagocytes transport oral antigens and promote immune tolerance, but their role in innate immune responses remains unclear. Here we found that intestinal phagocytes were anergic to ligands for Toll-like receptors (TLRs) or commensals but constitutively expressed the precursor to interleukin 1ß (pro-IL-1ß). After infection with pathogenic Salmonella or Pseudomonas, intestinal phagocytes produced mature IL-1ß through the NLRC4 inflammasome but did not produce tumor necrosis factor (TNF) or IL-6. BALB/c mice deficient in NLRC4 or the IL-1 receptor were highly susceptible to orogastric but not intraperitoneal infection with Salmonella. That enhanced lethality was preceded by impaired expression of endothelial adhesion molecules, lower neutrophil recruitment and poor intestinal pathogen clearance. Thus, NLRC4-dependent production of IL-1ß by intestinal phagocytes represents a specific response that discriminates pathogenic bacteria from commensal bacteria and contributes to host defense in the intestine.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Clonal Anergy , Host-Pathogen Interactions/immunology , Interleukin-1beta/metabolism , Intestines/immunology , Intestines/microbiology , Phagocytes/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Caspase 1/metabolism , Flagellin/immunology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-6/biosynthesis , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Phagocytes/microbiology , Pseudomonas/immunology , Pseudomonas Infections/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Salmonella/genetics , Salmonella/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
3.
Nat Immunol ; 11(10): 897-904, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20835230

ABSTRACT

Interleukin 1ß (IL-1ß) is an important inflammatory mediator of type 2 diabetes. Here we show that oligomers of islet amyloid polypeptide (IAPP), a protein that forms amyloid deposits in the pancreas during type 2 diabetes, triggered the NLRP3 inflammasome and generated mature IL-1ß. One therapy for type 2 diabetes, glyburide, suppressed IAPP-mediated IL-1ß production in vitro. Processing of IL-1ß initiated by IAPP first required priming, a process that involved glucose metabolism and was facilitated by minimally oxidized low-density lipoprotein. Finally, mice transgenic for human IAPP had more IL-1ß in pancreatic islets, which localized together with amyloid and macrophages. Our findings identify previously unknown mechanisms in the pathogenesis of type 2 diabetes and treatment of pathology caused by IAPP.


Subject(s)
Amyloid/metabolism , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/immunology , Interleukin-1beta/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diabetes Mellitus, Type 2/metabolism , Glyburide/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Islet Amyloid Polypeptide , Islets of Langerhans/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Rats , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics
4.
Nat Immunol ; 10(3): 241-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19221555

ABSTRACT

The inflammasome is a multiprotein complex that mediates the activation of caspase-1, which promotes secretion of the proinflammatory cytokines interleukin 1beta (IL-1beta) and IL-18, as well as 'pyroptosis', a form of cell death induced by bacterial pathogens. Members of the Nod-like receptor family, including NLRP1, NLRP3 and NLRC4, and the adaptor ASC are critical components of the inflammasome that link microbial and endogenous 'danger' signals to caspase-1 activation. Several diseases are associated with dysregulated activation of caspase-1 and secretion of IL-1beta. Thus, understanding inflammasome pathways may provide insight into disease pathogenesis that might identify potential targets for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Apoptosis Regulatory Proteins/immunology , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Carrier Proteins/immunology , Caspase 1/immunology , Interleukin-1beta/immunology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Humans , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Protein Interaction Domains and Motifs , Uric Acid/immunology , Uric Acid/metabolism
5.
Immunity ; 37(1): 85-95, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22819042

ABSTRACT

Cryopyrin-associated periodic syndromes (CAPS) are caused by aberrant interleukin-1ß (IL-1ß) production induced by mutations in the NLRP3 protein in humans, but the mechanisms involved remain poorly understood. Using a mouse model, we show a role for the indigenous microbiota and mast cells (MCs) in skin disease associated with mutant Nlrp3 protein. Unlike normal cells, MCs expressing mutant Nlrp3 produced IL-1ß in response to lipopolysaccharide or tumor necrosis factor-α (TNF-α). In neonatal mice, the microbiota induced TNF-α and IL-1ß and promoted skin disease. MC deficiency greatly reduced disease in Nlrp3 mutant mice, and reconstitution of MC-deficient mice with mutant MCs restored skin disease, which required the expression of IL-1ß in MCs. Surprisingly, neutralization of TNF-α abrogated IL-1ß production and skin disease in neonatal Nlrp3 mutant mice, but not in affected adult mice. Thus, the microbiota and MCs initiate cellular events leading to dysregulated IL-1ß production and skin inflammation in neonatal mice with the CAPS-associated Nlrp3 mutation.


Subject(s)
Carrier Proteins/genetics , Dermatitis/genetics , Dermatitis/immunology , Interleukin-1beta/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mutation , Animals , Caspase 1/metabolism , Dermatitis/microbiology , Female , Interleukin-1beta/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Skin Diseases/chemically induced , Skin Diseases/genetics , Skin Diseases/immunology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/immunology
6.
Immunity ; 34(5): 695-7, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21616439

ABSTRACT

Familial Mediterranean fever is caused by mutations of the PYRIN protein. Chae et al. (2011) provide evidence for a ASC protein-dependent pathway of caspase-1 activation in which gain-of-function PYRIN mutations lead to IL-1ß cytokine overproduction and inflammatory disease.

7.
Immunity ; 34(5): 769-80, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21565531

ABSTRACT

The intracellular sensor Nod2 is activated in response to bacteria, and the impairment of this response is linked to Crohn's disease. However, the function of Nod2 in host defense remains poorly understood. We found that Nod2-/- mice exhibited impaired intestinal clearance of Citrobacter rodentium, an enteric bacterium that models human infection by pathogenic Escherichia coli. The increased bacterial burden was preceded by reduced CCL2 chemokine production, inflammatory monocyte recruitment, and Th1 cell responses in the intestine. Colonic stromal cells, but not epithelial cells or resident CD11b+ phagocytic cells, produced CCL2 in response to C. rodentium in a Nod2-dependent manner. Unlike resident phagocytic cells, inflammatory monocytes produced IL-12, a cytokine that induces adaptive immunity required for pathogen clearance. Adoptive transfer of Ly6C(hi) monocytes restored the clearance of the pathogen in infected Ccr2-/- mice. Thus, Nod2 mediates CCL2-CCR2-dependent recruitment of inflammatory monocytes, which is important in promoting bacterial eradication in the intestine.


Subject(s)
Chemokine CCL2/immunology , Citrobacter rodentium/immunology , Colitis/immunology , Enterobacteriaceae Infections/immunology , Monocytes/immunology , Nod2 Signaling Adaptor Protein/immunology , Animals , Colitis/microbiology , Colitis/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/biosynthesis , Nod2 Signaling Adaptor Protein/deficiency , Stromal Cells/immunology
8.
J Immunol ; 198(7): 2735-2746, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28242647

ABSTRACT

Integration of signaling and metabolic pathways enables and sustains lymphocyte function. Whereas metabolic changes occurring during T cell activation are well characterized, the metabolic demands of differentiated T lymphocytes are largely unexplored. In this study, we defined the bioenergetics of Th17 effector cells generated in vivo. These cells depend on oxidative phosphorylation (OXPHOS) for energy and cytokine production. Mechanistically, the essential role of OXPHOS in Th17 cells results from their limited capacity to increase glycolysis in response to metabolic stresses. This metabolic program is observed in mouse and human Th17 cells, including those isolated from Crohn disease patients, and it is linked to disease, as inhibiting OXPHOS reduces the severity of murine colitis and psoriasis. These studies highlight the importance of analyzing metabolism in effector lymphocytes within in vivo inflammatory contexts and suggest a therapeutic role for manipulating OXPHOS in Th17-driven diseases.


Subject(s)
Cell Differentiation/immunology , Colitis/immunology , Lymphocyte Activation/immunology , Oxidative Phosphorylation , Th17 Cells/immunology , Animals , Cell Separation , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Transcriptome
9.
J Immunol ; 194(12): 5789-800, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25972478

ABSTRACT

The coinhibitory receptor programmed death-1 (PD-1) maintains immune homeostasis by negatively regulating T cell function and survival. Blockade of PD-1 increases the severity of graft-versus-host disease (GVHD), but the interplay between PD-1 inhibition and T cell metabolism is not well studied. We found that both murine and human alloreactive T cells concomitantly upregulated PD-1 expression and increased levels of reactive oxygen species (ROS) following allogeneic bone marrow transplantation. This PD-1(Hi)ROS(Hi) phenotype was specific to alloreactive T cells and was not observed in syngeneic T cells during homeostatic proliferation. Blockade of PD-1 signaling decreased both mitochondrial H2O2 and total cellular ROS levels, and PD-1-driven increases in ROS were dependent upon the oxidation of fatty acids, because treatment with etomoxir nullified changes in ROS levels following PD-1 blockade. Downstream of PD-1, elevated ROS levels impaired T cell survival in a process reversed by antioxidants. Furthermore, PD-1-driven changes in ROS were fundamental to establishing a cell's susceptibility to subsequent metabolic inhibition, because blockade of PD-1 decreased the efficacy of later F1F0-ATP synthase modulation. These data indicate that PD-1 facilitates apoptosis in alloreactive T cells by increasing ROS in a process dependent upon the oxidation of fat. In addition, blockade of PD-1 undermines the potential for subsequent metabolic inhibition, an important consideration given the increasing use of anti-PD-1 therapies in the clinic.


Subject(s)
Cell Survival , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens/immunology , Apoptosis/genetics , Apoptosis/immunology , Bone Marrow Transplantation/adverse effects , Cell Survival/genetics , Fatty Acids/metabolism , Female , Gene Expression , Graft vs Host Disease/etiology , Heterografts , Humans , Mice , Mice, Transgenic , Oxidation-Reduction , Phenotype , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Reactive Oxygen Species/metabolism
10.
Proc Natl Acad Sci U S A ; 111(40): E4254-63, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25246571

ABSTRACT

When nucleotide-binding oligomerization domain-like receptors (NLRs) sense cytosolic-invading bacteria, they induce the formation of inflammasomes and initiate an innate immune response. In quiescent cells, inflammasome activity is tightly regulated to prevent excess inflammation and cell death. Many bacterial pathogens provoke inflammasome activity and induce inflammatory responses, including cell death, by delivering type III secreted effectors, the rod component flagellin, and toxins. Recent studies indicated that Shigella deploy multiple mechanisms to stimulate NLR inflammasomes through type III secretion during infection. Here, we show that Shigella induces rapid macrophage cell death by delivering the invasion plasmid antigen H7.8 (IpaH7.8) enzyme 3 (E3) ubiquitin ligase effector via the type III secretion system, thereby activating the NLR family pyrin domain-containing 3 (NLRP3) and NLR family CARD domain-containing 4 (NLRC4) inflammasomes and caspase-1 and leading to macrophage cell death in an IpaH7.8 E3 ligase-dependent manner. Mice infected with Shigella possessing IpaH7.8, but not with Shigella possessing an IpaH7.8 E3 ligase-null mutant, exhibited enhanced bacterial multiplication. We defined glomulin/flagellar-associated protein 68 (GLMN) as an IpaH7.8 target involved in IpaH7.8 E3 ligase-dependent inflammasome activation. This protein originally was identified through its association with glomuvenous malformations and more recently was described as a member of a Cullin ring ligase inhibitor. Modifying GLMN levels through overexpression or knockdown led to reduced or augmented inflammasome activation, respectively. Macrophages stimulated with lipopolysaccharide/ATP induced GLMN puncta that localized with the active form of caspase-1. Macrophages from GLMN(+/-) mice were more responsive to inflammasome activation than those from GLMN(+/+) mice. Together, these results highlight a unique bacterial adaptation that hijacks inflammasome activation via interactions between IpaH7.8 and GLMN.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Muscle Proteins/metabolism , Shigella flexneri/metabolism , Animals , Antigens, Bacterial/genetics , Apoptosis , Bacterial Proteins/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Immunoblotting , Jurkat Cells , Macrophages/microbiology , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Fluorescence , Muscle Proteins/genetics , Protein Binding , Shigella flexneri/genetics , Shigella flexneri/physiology , Two-Hybrid System Techniques
11.
Biochim Biophys Acta ; 1847(11): 1469-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25979236

ABSTRACT

Mitochondria are multifunctional organelles that play a central role in cellular homeostasis. Severe mitochondrial dysfunction leads to life-threatening diseases in humans and accelerates the aging process. Surprisingly, moderate reduction of mitochondrial function in different species has anti-aging effects. High-throughput screenings in the nematode Caenorhabditis elegans lead to the identification of several pro-longevity genetic and pharmacological interventions. Large-scale screens, however, are manual, subjective, time consuming and costly. These limitations could be reduced by the identification of automatically quantifiable biomarkers of healthy aging. In this study we exploit the distinct and reproducible phenotypes described in C. elegans upon different levels of mitochondrial alteration to develop an automated high-content strategy to identify new potential pro-longevity interventions. Utilizing the microscopy platform Cellomics ArrayScan Reader, we optimize a workflow to automatically and reliably quantify the discrete phenotypic readouts associated with different degrees of silencing of mitochondrial respiratory chain regulatory proteins, and validate the approach with mitochondrial-targeting drugs known to extend lifespan in C. elegans. Finally, we report that a new mitochondrial ATPase modulator matches our screening phenotypic criteria and extends nematode's lifespan thus providing the proof of principle that our strategy could be exploited to identify novel mitochondrial-targeted drugs with pro-longevity activity. This article is part of a Special Issue entitled: Mitochondrial Dysfunction in Aging.


Subject(s)
Caenorhabditis elegans/physiology , Longevity , Mitochondria/physiology , Animals , Microscopy , Mitochondria/drug effects , Phenotype
12.
PLoS Pathog ; 10(2): e1003926, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24516390

ABSTRACT

Recognition of intracellular pathogenic bacteria by members of the nucleotide-binding domain and leucine-rich repeat containing (NLR) family triggers immune responses against bacterial infection. A major response induced by several Gram-negative bacteria is the activation of caspase-1 via the Nlrc4 inflammasome. Upon activation, caspase-1 regulates the processing of proIL-1ß and proIL-18 leading to the release of mature IL-1ß and IL-18, and induction of pyroptosis. The activation of the Nlrc4 inflammasome requires the presence of an intact type III or IV secretion system that mediates the translocation of small amounts of flagellin or PrgJ-like rod proteins into the host cytosol to induce Nlrc4 activation. Using the Salmonella system, it was shown that Naip2 and Naip5 link flagellin and the rod protein PrgJ, respectively, to Nlrc4. Furthermore, phosphorylation of Nlrc4 at Ser533 by Pkcδ was found to be critical for the activation of the Nlrc4 inflammasome. Here, we show that Naip2 recognizes the Shigella T3SS inner rod protein MxiI and induces Nlrc4 inflammasome activation. The expression of MxiI in primary macrophages was sufficient to induce pyroptosis and IL-1ß release, which were prevented in macrophages deficient in Nlrc4. In the presence of MxiI or Shigella infection, MxiI associated with Naip2, and Naip2 interacted with Nlrc4. siRNA-mediated knockdown of Naip2, but not Naip5, inhibited Shigella-induced caspase-1 activation, IL-1ß maturation and Asc pyroptosome formation. Notably, the Pkcδ kinase was dispensable for caspase-1 activation and secretion of IL-1ß induced by Shigella or Salmonella infection. These results indicate that activation of caspase-1 by Shigella is triggered by the rod protein MxiI that interacts with Naip2 to induce activation of the Nlrc4 inflammasome independently of the Pkcδ kinase.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/metabolism , Calcium-Binding Proteins/metabolism , Host-Parasite Interactions/immunology , Inflammasomes/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Protein Kinase C-delta/metabolism , Animals , Caspase 1/metabolism , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Transfection
13.
J Immunol ; 193(8): 4214-4222, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25225670

ABSTRACT

The nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (Nlrp3) inflammasome plays an important role in inflammation by controlling the maturation and secretion of the cytokines IL-1ß and IL-18 in response to multiple stimuli including pore-forming toxins, particulate matter, and ATP. Although the pathways activated by the latter stimuli lead to a decrease in intracellular K(+) concentration, which is required for inflammasome activation, the mechanism by which microbial RNA activates Nlrp3, remains poorly understood. In this study, we found that cytosolic poly(I:C), but not total RNA from healthy macrophages, macrophages undergoing pyroptosis, or mitochondrial RNA, induces caspase-1 activation and IL-1ß release through the Nlrp3 inflammasome. Experiments with macrophages deficient in Tlr3, Myd88, or Trif, indicate that poly(I:C) induces Nlrp3 activation independently of TLR signaling. Further analyses revealed that the cytosolic sensors Rig-I and melanoma differentiation-associated gene 5 act redundantly via the common adaptor mitochondrial antiviral signaling (Mavs) to induce Nlrp3 activation in response to poly(I:C), but not ATP or nigericin. Mechanistically, Mavs triggered membrane permeabilization and K(+) efflux independently of the inflammasome which were required for poly(I:C)-induced Nlrp3 activation. We conclude that poly (I:C) activates the inflammasome through an Mavs-dependent surveillance pathway that converges into a common K(+) lowering step in the cytosol that is essential for the induction of Nlrp3 activation.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Carrier Proteins/immunology , Potassium/metabolism , RNA, Double-Stranded/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Caspase 1/immunology , Cytosol , DEAD Box Protein 58 , DEAD-box RNA Helicases/immunology , Inflammation/immunology , Interferon-Induced Helicase, IFIH1 , Interleukin-18/biosynthesis , Interleukin-18/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Ion Transport , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Poly I-C/immunology , RNA, Bacterial/immunology , RNA, Viral/immunology , Signal Transduction/immunology , Toll-Like Receptor 3/genetics
14.
Nature ; 464(7293): 1357-61, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20428172

ABSTRACT

The inflammatory nature of atherosclerosis is well established but the agent(s) that incite inflammation in the artery wall remain largely unknown. Germ-free animals are susceptible to atherosclerosis, suggesting that endogenous substances initiate the inflammation. Mature atherosclerotic lesions contain macroscopic deposits of cholesterol crystals in the necrotic core, but their appearance late in atherogenesis had been thought to disqualify them as primary inflammatory stimuli. However, using a new microscopic technique, we revealed that minute cholesterol crystals are present in early diet-induced atherosclerotic lesions and that their appearance in mice coincides with the first appearance of inflammatory cells. Other crystalline substances can induce inflammation by stimulating the caspase-1-activating NLRP3 (NALP3 or cryopyrin) inflammasome, which results in cleavage and secretion of interleukin (IL)-1 family cytokines. Here we show that cholesterol crystals activate the NLRP3 inflammasome in phagocytes in vitro in a process that involves phagolysosomal damage. Similarly, when injected intraperitoneally, cholesterol crystals induce acute inflammation, which is impaired in mice deficient in components of the NLRP3 inflammasome, cathepsin B, cathepsin L or IL-1 molecules. Moreover, when mice deficient in low-density lipoprotein receptor (LDLR) were bone-marrow transplanted with NLRP3-deficient, ASC (also known as PYCARD)-deficient or IL-1alpha/beta-deficient bone marrow and fed on a high-cholesterol diet, they had markedly decreased early atherosclerosis and inflammasome-dependent IL-18 levels. Minimally modified LDL can lead to cholesterol crystallization concomitant with NLRP3 inflammasome priming and activation in macrophages. Although there is the possibility that oxidized LDL activates the NLRP3 inflammasome in vivo, our results demonstrate that crystalline cholesterol acts as an endogenous danger signal and its deposition in arteries or elsewhere is an early cause rather than a late consequence of inflammation. These findings provide new insights into the pathogenesis of atherosclerosis and indicate new potential molecular targets for the therapy of this disease.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Carrier Proteins/metabolism , Cholesterol/chemistry , Cholesterol/metabolism , Animals , Apoptosis Regulatory Proteins , Atherosclerosis/chemically induced , Bone Marrow Transplantation , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Cathepsin B/metabolism , Cathepsin L/metabolism , Cholesterol/pharmacology , Crystallization , Cytoskeletal Proteins/deficiency , Diet, Atherogenic , Female , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Interleukin-1/deficiency , Interleukin-18/metabolism , Lysosomes/drug effects , Lysosomes/pathology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Peritoneal Cavity/pathology , Phagocytes/drug effects , Phagocytes/pathology , Phagocytes/physiology , Receptors, LDL/deficiency , Time Factors
15.
J Immunol ; 190(1): 334-9, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225887

ABSTRACT

On the basis of studies in mouse macrophages, activation of the nucleotide-binding oligomerization domain-like receptor (NLR) pyrin domain-containing 3 (Nlrp3) inflammasome is thought to require two signals. The first signal is provided by TLR stimulation and triggers the synthesis of the IL-1ß precursor and Nlrp3. The second signal can be mediated by stimulation of the purinergic receptor P2X ligand-gated ion channel 7 (P2X7) by millimolar concentrations of ATP. However, these high concentrations of ATP are not found normally in the in vivo extracellular milieu, raising concern about the physiological relevance of the ATP-P2X7 pathway of inflammasome activation. In this article, we show that unlike macrophages, murine bone marrow-derived and splenic dendritic cells (DCs) can secrete substantial amounts of mature IL-1ß upon stimulation with TLR ligands in the absence of ATP stimulation. The differential ability of DCs to release IL-1ß and activate caspase-1 was associated with increased expression of Nlrp3 under steady-state conditions and of pro-IL-1ß and Nlrp3 after stimulation with TLR agonists. IL-1ß secretion from stimulated DCs was largely dependent on the Nlrp3 inflammasome, but independent of P2X7 and unaffected by incubation with apyrase. More importantly, i.p. administration of LPS induced IL-1ß production in serum, which was abrogated in Nlrp3-null mice but was unaffected in P2X7-deficient mice. These results demonstrate differential regulation of the Nlrp3 inflammasome in macrophages and DCs. Furthermore, they challenge the idea that the ATP-P2X7 axis is critical for TLR-induced IL-1ß production via the Nlrp3 inflammasome in vivo.


Subject(s)
Carrier Proteins/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interleukin-1beta/biosynthesis , Receptors, Purinergic P2X7/physiology , Toll-Like Receptors/agonists , Adenosine Triphosphate/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Dendritic Cells/pathology , Inflammasomes/metabolism , Inflammasomes/physiology , Interleukin-1beta/metabolism , Interleukin-1beta/physiology , Ligands , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Precursors/biosynthesis , Protein Precursors/metabolism , Receptors, Purinergic P2X7/deficiency , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Toll-Like Receptors/metabolism
16.
Antimicrob Agents Chemother ; 58(6): 3043-52, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24614386

ABSTRACT

We report here the synthesis of 2-aminothiazolones along with their biological properties as novel anti-HIV agents. Such compounds have proven to act through the inhibition of the gp120-CD4 protein-protein interaction that occurs at the very early stage of the HIV-1 entry process. No cytotoxicity was found for these compounds, and broad antiviral activities against laboratory strains and pseudotyped viruses were documented. Docking simulations have also been applied to predict the mechanism, at the molecular level, by which the inhibitors were able to interact within the Phe43 cavity of HIV-1 gp120. Furthermore, a preliminary absorption, distribution, metabolism, and excretion (ADME) evaluation was performed. Overall, this study led the basis for the development of more potent HIV entry inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , CD4 Antigens/drug effects , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Anti-HIV Agents/chemistry , CD4 Antigens/chemistry , CD4 Antigens/metabolism , Cell Line , HIV Envelope Protein gp120/metabolism , HIV Fusion Inhibitors/chemistry , Humans , Molecular Docking Simulation , Protein Binding
17.
Eur J Immunol ; 43(5): 1147-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23401008

ABSTRACT

Inflammasomes are multi-protein platforms that drive the activation of caspase-1 leading to the processing and secretion of biologically active IL-1ß and IL-18. Different inflammasomes including NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), NLR caspase-recruitment domain-containing 4 (NLRC4) and absent in melanoma 2 (AIM2) are activated and assembled in response to distinct microbial or endogenous stimuli. However, the mechanisms by which upstream stimuli trigger inflammasome activation remain poorly understood. Double-stranded RNA-activated protein kinase (PKR), a protein kinase activated by viral infection, has been recently shown to be required for the activation of the inflammasomes. Using macrophages from two different mouse strains deficient in PKR, we found that PKR is important for the induction of the inducible nitric oxide synthase (iNOS). However, PKR was dispensable for caspase-1 activation, processing of pro-IL-1ß/IL-18 and secretion of IL-1ß induced by stimuli that trigger the activation of NLRP3, NLRC4 and AIM2. These results indicate that PKR is not required for inflammasome activation in macrophages.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/drug effects , Nitric Oxide Synthase Type II/immunology , eIF-2 Kinase/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Caspase 1/genetics , Caspase 1/immunology , Cells, Cultured , DNA-Binding Proteins , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-18/immunology , Interleukin-18/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nitric Oxide Synthase Type II/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Protein Precursors/immunology , Protein Precursors/metabolism , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
18.
Eur J Immunol ; 43(10): 2650-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23818043

ABSTRACT

Helicobacter pylori colonization of the stomach affects about half of the world population and is associated with the development of gastritis, ulcers, and cancer. Polymorphisms in the IL1B gene are linked to an increased risk of H. pylori associated cancer, but the bacterial and host factors that regulate interleukin (IL)-1ß production in response to H. pylori infection remain unknown. Using murine BM-derived DCs, we show that the bacterial virulence factors cytotoxin-associated genes pathogenicity island and CagL, but not vacuolating cytotoxin A or CagA, regulate the induction of pro-IL-1ß and the production of mature IL-1ß in response to H. pylori infection. We further show that the host receptors, Toll-like receptor 2 (TLR2) and nucleotide-binding oligomerization domain 2 (NOD2), but not NOD1, are required for induction of pro-IL-1ß and NOD-like receptor pyrin domain containing 3 (NLRP3) in H. pylori infected DCs. In contrast, NLRP3 and the adaptor ASC were essential for the activation of caspase-1, processing of pro-IL-1ß into IL-1ß, and IL-1ß secretion. Finally, we show that mice deficient in caspase-1, IL-1ß, and IL-1 receptor, but not NLRP3, are impaired in the clearance of CagA-positive H. pylori from the stomach when compared with WT mice. These studies identify bacterial cag pathogenicity island and the cooperative interaction among host innate receptors TLR2, NOD2, and NLRP3 as important regulators of IL-1ß production in H. pylori infected DCs.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Dendritic Cells/immunology , Genomic Islands , Helicobacter Infections/immunology , Helicobacter pylori/pathogenicity , Nod2 Signaling Adaptor Protein/metabolism , Toll-Like Receptor 2/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Load , Carrier Proteins/genetics , Dendritic Cells/microbiology , Gene Expression Regulation/genetics , Immunity, Innate/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nod2 Signaling Adaptor Protein/genetics , Toll-Like Receptor 2/genetics , Virulence/genetics
19.
PLoS Pathog ; 8(7): e1002628, 2012.
Article in English | MEDLINE | ID: mdl-22807671

ABSTRACT

The cysteine protease caspase-7 has an established role in the execution of apoptotic cell death, but recent findings also suggest involvement of caspase-7 during the host response to microbial infection. Caspase-7 can be cleaved by the inflammatory caspase, caspase-1, and has been implicated in processing and activation of microbial virulence factors. Thus, caspase-7 function during microbial infection may be complex, and its role in infection and immunity has yet to be fully elucidated. Here we demonstrate that caspase-7 is cleaved during cytosolic infection with the intracellular bacterial pathogen, Listeria monocytogenes. Cleavage of caspase-7 during L. monocytogenes infection did not require caspase-1 or key adaptors of the primary pathways of innate immune signaling in this infection, ASC, RIP2 and MyD88. Caspase-7 protected infected macrophages against plasma membrane damage attributable to the bacterial pore-forming toxin Listeriolysin O (LLO). LLO-mediated membrane damage could itself trigger caspase-7 cleavage, independently of infection or overt cell death. We also detected caspase-7 cleavage upon treatment with other bacterial pore-forming toxins, but not in response to detergents. Taken together, our results support a model where cleavage of caspase-7 is a consequence of toxin-mediated membrane damage, a common occurrence during infection. We propose that host activation of caspase-7 in response to pore formation represents an adaptive mechanism by which host cells can protect membrane integrity during infection.


Subject(s)
Caspase 7/metabolism , Cell Membrane/microbiology , Listeria monocytogenes/pathogenicity , Listeriosis/enzymology , Listeriosis/pathology , Macrophages/metabolism , Animals , Apoptosis Regulatory Proteins , Bacterial Toxins/metabolism , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Cells, Cultured , Cytoskeletal Proteins/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Listeria monocytogenes/immunology , Listeria monocytogenes/metabolism , Listeriosis/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Virulence Factors/metabolism
20.
Int J Med Microbiol ; 304(3-4): 384-92, 2014 May.
Article in English | MEDLINE | ID: mdl-24581881

ABSTRACT

Crohn's disease (CD) is a multifactorial pathology associated with the presence of adherent-invasive Escherichia coli (AIEC) and NLRP3 polymorphic variants. The presence of intracellular E. coli in other intestinal pathologies (OIP) and the role of NLRP3-inflammasome in the immune response activated by these bacteria have not been investigated. In this study, we sought to characterize intracellular strains isolated from patients with CD, ulcerative colitis (UC) and OIP, and analyze NLRP3-inflammasome role in the immune response and bactericidal activity induced in macrophages exposed to invasive bacteria. For this, intracellular E. coli isolation from ileal biopsies, using gentamicin-protection assay, revealed a prevalence and CFU/biopsy of E. coli higher in biopsies from CD, UC and OIP patients than in controls. To characterize bacterial isolates, pulsed-field gel electrophoresis (PFGE) patterns, virulence genes, serogroup and phylogenetic group were analyzed. We found out that bacteria isolated from a given patient were closely related and shared virulence factors; however, strains from different patients were genetically heterogeneous. AIEC characteristics in isolated strains, such as invasive and replicative properties, were assessed in epithelial cells and macrophages, respectively. Some strains from CD and UC demonstrated AIEC properties, but not strains from OIP. Furthermore, the role of NLRP3 in pro-inflammatory cytokines production and bacterial elimination was determined in macrophages. E. coli strains induced IL-1ß through NLRP3-dependent mechanism; however, their elimination by macrophages was independent of NLRP3. Invasiveness of intracellular E. coli strains into the intestinal mucosa and IL-1ß production may contribute to CD and UC pathogenesis.


Subject(s)
Carrier Proteins/metabolism , Escherichia coli/physiology , Host-Pathogen Interactions , Inflammasomes/metabolism , Inflammatory Bowel Diseases/microbiology , Macrophages/microbiology , Microbial Viability , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Bacterial Load , Biopsy , Cell Line , Cytosol/microbiology , Epithelial Cells/microbiology , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli/isolation & purification , Female , Genotype , Humans , Ileum/microbiology , Ileum/pathology , Male , Mice , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein , Virulence Factors/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL