Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Inflammation ; 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38329636

ABSTRACT

Neutrophilic asthma is generally defined by poorly controlled symptoms and high levels of neutrophils in the lungs. Short-chain fatty acids (SCFAs) are proposed as nonpharmacological therapy for allergic asthma, but their impact on the neutrophilic asthma lacks evidence. SCFAs regulate immune cell responses and impact the inflammasome NLRP3, a potential pharmacological target for neutrophilic asthma. Here, we explored the capacity of SCFAs to mitigate murine-induced neutrophilic asthma and the contribution of NLRP3 to this asthma. The objective of this study is to analyze whether SCFAs can attenuate lung inflammation and tissue remodeling in murine neutrophilic asthma and NLRP3 contribution to this endotype. Wild-type (WT) C57BL6 mice orotracheally received 10 µg of HDM (house dust mite) in 80 µL of saline on days 0, 6-10. To explore SCFAs, each HDM group received 200 mM acetate, propionate, or butyrate. To explore NLRP3, Nlrp3 KO mice received the same protocol of HDM. On the 14th day, after euthanasia, bronchoalveolar lavage fluid (BALF) and lungs were collected to evaluate cellularity, inflammatory cytokines, and tissue remodeling. HDM group had increased BALF neutrophil influx, TNF-α, IFN-γ, IL-17A, collagen deposition, and mucus secretion compared to control. SCFAs distinctively attenuate lung inflammation. Only features of tissue remodeling were Nlrp3-dependent such as collagen deposition, mucus secretion, active TGF-ß cytokine, and IMs CD206+. SCFAs greatly decreased inflammatory cytokines and tissue remodeling. Only tissue remodeling was dependent on NLRP3. It reveals the potential of SCFAs to act as an additional therapy to mitigate neutrophilic asthma and the NLRP3 contribution to asthma.

3.
Cell Death Dis ; 12(7): 692, 2021 07 10.
Article in English | MEDLINE | ID: mdl-34247195

ABSTRACT

Chagas disease is a life-threatening disorder caused by the protozoan parasite Trypanosoma cruzi. Parasite-specific antibodies, CD8+ T cells, as well as IFN-γ and nitric oxide (NO) are key elements of the adaptive and innate immunity against the extracellular and intracellular forms of the parasite. Bim is a potent pro-apoptotic member of the Bcl-2 family implicated in different aspects of the immune regulation, such as negative selection of self-reactive thymocytes and elimination of antigen-specific T cells at the end of an immune response. Interestingly, the role of Bim during infections remains largely unidentified. To explore the role of Bim in Chagas disease, we infected WT, Bim+/-, Bim-/- mice with trypomastigotes forms of the Y strain of T. cruzi. Strikingly, our data revealed that Bim-/- mice exhibit a delay in the development of parasitemia followed by a deficiency in the control of parasite load in the bloodstream and a decreased survival compared to WT and Bim+/- mice. At the peak of parasitemia, peritoneal macrophages of Bim-/- mice exhibit decreased NO production, which correlated with a decrease in the pro-inflammatory Small Peritoneal Macrophage (SPM) subset. A similar reduction in NO secretion, as well as in the pro-inflammatory cytokines IFN-γ and IL-6, was also observed in Bim-/- splenocytes. Moreover, an impaired anti-T. cruzi CD8+ T-cell response was found in Bim-/- mice at this time point. Taken together, our results suggest that these alterations may contribute to the establishment of a delayed yet enlarged parasitic load observed at day 9 after infection of Bim-/- mice and place Bim as an important protein in the control of T. cruzi infections.


Subject(s)
Bcl-2-Like Protein 11/deficiency , Chagas Disease/parasitology , Trypanosoma cruzi/pathogenicity , Animals , Bcl-2-Like Protein 11/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/parasitology , Cells, Cultured , Chagas Disease/genetics , Chagas Disease/immunology , Chagas Disease/metabolism , Disease Models, Animal , Female , Host-Parasite Interactions , Interferon-gamma/metabolism , Interleukin-6/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/parasitology , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Parasite Load , Spleen/immunology , Spleen/metabolism , Spleen/parasitology , Time Factors , Trypanosoma cruzi/immunology
4.
Front Cell Infect Microbiol ; 11: 676183, 2021.
Article in English | MEDLINE | ID: mdl-34123875

ABSTRACT

Deficiency in memory formation and increased immunosenescence are pivotal features of Trypanosoma cruzi infection proposed to play a role in parasite persistence and disease development. The vaccination protocol that consists in a prime with plasmid DNA followed by the boost with a deficient recombinant human adenovirus type 5, both carrying the ASP2 gene of T. cruzi, is a powerful strategy to elicit effector memory CD8+ T-cells against this parasite. In virus infections, the inhibition of mTOR, a kinase involved in several biological processes, improves the response of memory CD8+ T-cells. Therefore, our aim was to assess the role of rapamycin, the pharmacological inhibitor of mTOR, in CD8+ T response against T. cruzi induced by heterologous prime-boost vaccine. For this purpose, C57BL/6 or A/Sn mice were immunized and daily treated with rapamycin for 34 days. CD8+ T-cells response was evaluated by immunophenotyping, intracellular staining, ELISpot assay and in vivo cytotoxicity. In comparison with vehicle-injection, rapamycin administration during immunization enhanced the frequency of ASP2-specific CD8+ T-cells and the percentage of the polyfunctional population, which degranulated (CD107a+) and secreted both interferon gamma (IFNγ) and tumor necrosis factor (TNF). The beneficial effects were long-lasting and could be detected 95 days after priming. Moreover, the effects were detected in mice immunized with ten-fold lower doses of plasmid/adenovirus. Additionally, the highly susceptible to T. cruzi infection A/Sn mice, when immunized with low vaccine doses, treated with rapamycin, and challenged with trypomastigote forms of the Y strain showed a survival rate of 100%, compared with 42% in vehicle-injected group. Trying to shed light on the biological mechanisms involved in these beneficial effects on CD8+ T-cells by mTOR inhibition after immunization, we showed that in vivo proliferation was higher after rapamycin treatment compared with vehicle-injected group. Taken together, our data provide a new approach to vaccine development against intracellular parasites, placing the mTOR inhibitor rapamycin as an adjuvant to improve effective CD8+ T-cell response.


Subject(s)
Protozoan Vaccines , Trypanosoma cruzi , Animals , CD8-Positive T-Lymphocytes , Mice , Mice, Inbred C57BL , Sirolimus/pharmacology , Vaccination
5.
Cells ; 10(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33440601

ABSTRACT

This study evaluated the role of endogenous and exogenous annexin A1 (AnxA1) in the activation of the NLRP3 inflammasome in isolated peritoneal neutrophils. C57BL/6 wild-type (WT) and AnxA1 knockout mice (AnxA1-/-) received 0.3% carrageenan intraperitoneally and, after 3 h, the peritoneal exudate was collected. WT and AnxA1-/- neutrophils were then stimulated with lipopolysaccharide, followed by the NLRP3 agonists nigericin or ATP. To determine the exogenous effect of AnxA1, the neutrophils were pretreated with the AnxA1-derived peptide Ac2-26 followed by the NLRP3 agonists. Ac2-26 administration reduced NLRP3-derived IL-1ß production by WT neutrophils after nigericin and ATP stimulation. However, IL-1ß release was impaired in AnxA1-/- neutrophils stimulated by both agonists, and there was no further impairment in IL-1ß release with Ac2-26 treatment before stimulation. Despite this, ATP- and nigericin-stimulated AnxA1-/- neutrophils had increased levels of cleaved caspase-1. The lipidomics of supernatants from nigericin-stimulated WT and AnxA1-/- neutrophils showed potential lipid biomarkers of cell stress and activation, including specific sphingolipids and glycerophospholipids. AnxA1 peptidomimetic treatment also increased the concentration of phosphatidylserines and oxidized phosphocholines, which are lipid biomarkers related to the inflammatory resolution pathway. Together, our results indicate that exogenous AnxA1 negatively regulates NLRP3-derived IL-1ß production by neutrophils, while endogenous AnxA1 is required for the activation of the NLRP3 machinery.


Subject(s)
Annexin A1/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/metabolism , Animals , Inflammasomes/ultrastructure , Interleukin-1beta/metabolism , Lipids/chemistry , Male , Mice, Inbred C57BL , Neutrophil Activation , Neutrophils/ultrastructure
6.
Nat Rev Immunol ; 20(10): 594-602, 2020 10.
Article in English | MEDLINE | ID: mdl-32913283
7.
Front Immunol ; 11: 536, 2020.
Article in English | MEDLINE | ID: mdl-32328060

ABSTRACT

Efficient induction of effector and long-term protective antigen-specific CD8+ T memory response by vaccination is essential to eliminate malignant and pathogen-infected cells. Intracellular infectious bacteria, including Listeria monocytogenes, have been considered potent vectors to carry multiple therapeutic proteins and generate antigen-specific CD8+ T cell responses. Although the role of molecules involved in inflammatory cell death pathways, such as necroptosis (RIPK3-mediated) and pyroptosis (Caspase-1/11-mediated), as effectors of immune response against intracellular bacteria are relatively well understood, their contribution to the adjuvant effect of recombinant bacterial vectors in the context of antigen-specific CD8+ T cell response remained obscure. Therefore, we evaluated the impact of RIPK3 and Caspase-1/11 (Casp-1/11) individual and combined deficiencies on the modulation of antigen-specific CD8+ T cell response during vaccination of mice with ovalbumin-expressing L. monocytogenes (LM-OVA). We observed that Casp-1/11 but not RIPK3 deficiency negatively impacts the capacity of mice to clear LM-OVA. Importantly, both RIPK3 and Casp-1/11 are necessary for optimal LM-OVA-mediated antigen-specific CD8+ T cell response, as measured by in vivo antigen-specific CD8+ T cell proliferation, target cell elimination, and cytokine production. Furthermore, Casp-1/11 and Casp-1/11/RIPK3 combined deficiencies restrict the early initiation of antigen-specific CD8+ T cell memory response. Taken together, our findings demonstrate that RIPK3 and Casp-1/11 influence the quality of CD8+ T cell responses induced by recombinant L. monocytogenes vectors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Caspase 1/immunology , Caspases, Initiator/immunology , Genetic Vectors/immunology , Listeria monocytogenes/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL
8.
Sci Adv ; 6(10): eaax6346, 2020 03.
Article in English | MEDLINE | ID: mdl-32181339

ABSTRACT

Placental malaria (PM) is associated with severe inflammation leading to abortion, preterm delivery, and intrauterine growth restriction. Innate immunity responses play critical roles, but the mechanisms underlying placental immunopathology are still unclear. Here, we investigated the role of inflammasome activation in PM by scrutinizing human placenta samples from an endemic area and ablating inflammasome components in a PM mouse model. The reduction in birth weight in babies from infected mothers is paralleled by increased placental expression of AIM2 and NLRP3 inflammasomes. Using genetic dissection, we reveal that inflammasome activation pathways are involved in the production and detrimental action of interleukin-1ß (IL-1ß) in the infected placenta. The IL-1R pharmacological antagonist Anakinra improved pregnancy outcomes by restoring fetal growth and reducing resorption in an experimental model. These findings unveil that IL-1ß-mediated signaling is a determinant of PM pathogenesis, suggesting that IL-1R antagonists can improve clinical outcomes of malaria infection in pregnancy.


Subject(s)
Inflammasomes/drug effects , Interleukin-1beta/immunology , Malaria, Falciparum/immunology , Malaria/immunology , Plasmodium falciparum/pathogenicity , Pregnancy Complications, Parasitic/immunology , Signal Transduction/drug effects , Animals , Caspase 1/genetics , Caspase 1/immunology , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Female , Gene Expression Regulation , Humans , Immunity, Innate , Immunologic Factors/pharmacology , Inflammasomes/genetics , Inflammasomes/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Malaria/drug therapy , Malaria/genetics , Malaria/parasitology , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Plasmodium berghei/immunology , Plasmodium berghei/pathogenicity , Plasmodium falciparum/immunology , Pregnancy , Pregnancy Complications, Parasitic/genetics , Pregnancy Complications, Parasitic/parasitology , Pregnancy Complications, Parasitic/prevention & control , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Signal Transduction/immunology , THP-1 Cells , Trophoblasts/drug effects , Trophoblasts/immunology , Trophoblasts/parasitology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
J Leukoc Biol ; 106(3): 531-540, 2019 09.
Article in English | MEDLINE | ID: mdl-31071239

ABSTRACT

Autophagy and inflammasome activation are cell-autonomous and cross-regulated processes involved in host resistance against infections. Our group previously described that NLRP3 inflammasome is required for the control of Trypanosoma cruzi, the causative agent of Chagas disease. However, the involvement of autophagy in this process was unclear. Here, we demonstrated that T. cruzi was able to induce an increase in LC3-II expression as well as autophagosome and autolysosome formation in peritoneal macrophages (PMs) from C57BL/6 wild-type mice. Moreover, the pharmacologic inhibition of autophagic machinery impaired the ability of PMs to control T. cruzi replication. Importantly, NLRP3 was required for the induction of a regular autophagic flux in response to T. cruzi, an effect mediated by its participation in the autolysosomes formation. Together, these results indicate autophagy as an effector mechanism mediated by NLRP3 to control T. cruzi infection.


Subject(s)
Autophagy , Chagas Disease/metabolism , Chagas Disease/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acids/metabolism , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Lysosomes/metabolism , Lysosomes/ultrastructure , Macrophages/metabolism , Macrophages/parasitology , Mice, Inbred C57BL , Trypanosoma cruzi
10.
J Leukoc Biol ; 106(1): 201-207, 2019 07.
Article in English | MEDLINE | ID: mdl-30997938

ABSTRACT

Trypanossoma cruzi (T. cruzi), the causative protozoan of Chagas disease (CD) invades many cell types, including central nervous system (CNS) cells triggering local lesions and neurological impact. Previous work from our group described NLRP3 inflammasomes as central effectors for the parasite control by macrophages. Recent evidences demonstrate that NLRP3 can be activated in CNS cells with controversial consequences to the control of infections and inflammatory pathologies. However, the relative contribution of NLRP3 in different cell types remains to be elucidated. In this article, we described an effector response mediated by NLRP3 that works on microglia but not on astrocytes to control T. cruzi infection. Despite T. cruzi ability to invade astrocytes and microglia, astrocytes were clearly more permissive to parasite replication. Moreover, the absence of NLRP3 renders microglia but not astrocytes more permissive to T. cruzi replication. In fact, microglia but not astrocytes were able to secrete NLRP3-dependent IL-1ß and NO in response to T. cruzi. Importantly, the pharmacological inhibition of iNOS with aminoguanidine resulted in a significant increase in the numbers of amastigotes found in microglia from wild-type but not from NLRP3-/- mice, indicating the importance of NLRP3-mediated NO secretion to the infection control by these cells. Taken together, our findings revealed that T. cruzi differentially activates NLRP3 inflammasomes in astrocytes and microglia and established a role for these platforms in the control of a protozoan infection by glial cells from CNS.


Subject(s)
Astrocytes/parasitology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Nitric Oxide/biosynthesis , Trypanosoma cruzi/physiology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Microglia/parasitology
11.
Front Immunol ; 9: 2379, 2018.
Article in English | MEDLINE | ID: mdl-30459758

ABSTRACT

Pattern Recognition Receptors (PRRs) are proteins capable of recognizing molecules frequently found in pathogens (the so-called Pathogen-Associated Molecular Patterns-PAMPs), or molecules released by damaged cells (the Damage-Associated Molecular Patterns-DAMPs). They emerged phylogenetically prior to the appearance of the adaptive immunity and, therefore, are considered part of the innate immune system. Signals derived from the engagement of PRRs on the immune cells activate microbicidal and pro-inflammatory responses required to eliminate or, at least, to contain infectious agents. Molecularly controlled forms of cell death are also part of a very ancestral mechanism involved in key aspects of the physiology of multicellular organism, including the elimination of unwanted, damaged or infected cells. Interestingly, each form of cell death has its particular effect on inflammation and on the development of innate and adaptive immune responses. In this review article, we discuss some aspects of the molecular interplay between the cell death machinery and signals initiated by the activation of PRRs by PAMPs and DAMPs.


Subject(s)
Pathogen-Associated Molecular Pattern Molecules/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Apoptosis , Cell Death , Host-Pathogen Interactions , Humans , Inflammation/etiology , Inflammation/metabolism , Pyroptosis
12.
Sci Rep ; 7: 41686, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28150715

ABSTRACT

Nitric oxide synthase 2, inducible (Nos2) expression is necessary for the microbicidal activity of macrophages. However, NOS2 over-activation causes multiple inflammatory disorders, suggesting a tight gene regulation is necessary. Using cytosolic flagellin as a model for inflammasome-dependent NOS2 activation, we discovered a surprising new role for NLRC4/caspase-1 axis in regulating chromatin accessibility of the Nos2 promoter. We found that activation of two independent mechanisms is necessary for NOS2 expression by cytosolic flagellin: caspase-1 and NF-κB activation. NF-κB activation was necessary, but not sufficient, for NOS2 expression. Conversely, caspase-1 was necessary for NOS2 expression, but dispensable for NF-κB activation, indicating that this protease acts downstream NF-κB activation. We demonstrated that epigenetic regulation of Nos2 by caspase-1 involves cleavage of the chromatin regulator PARP1 (also known as ARTD1) and chromatin accessibility of the NF-κB binding sites located at the Nos2 promoter. Remarkably, caspase-1-mediated Nos2 transcription and NO production contribute to the resistance of macrophages to Salmonella typhimurium infection. Our results uncover the molecular mechanism behind the constricted regulation of Nos2 expression and open new therapeutic opportunities based on epigenetic activities of caspase-1 against infectious and inflammatory diseases.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Epigenesis, Genetic , Inflammasomes/metabolism , Nitric Oxide Synthase Type II/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Animals , Base Sequence , Binding Sites , Caspase 1/metabolism , Cytosol/metabolism , Flagellin/metabolism , Gene Expression Regulation , Mice , Mice, Knockout , Models, Biological , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Nucleotide Motifs , Promoter Regions, Genetic , Protein Binding , Proteolysis
13.
Mediators Inflamm ; 2014: 912965, 2014.
Article in English | MEDLINE | ID: mdl-25104883

ABSTRACT

Over the past 20 years, the immune effector mechanisms involved in the control of Trypanosoma cruzi, as well as the receptors participating in parasite recognition by cells of the innate immune system, have been largely described. However, the main questions on the physiopathology of Chagas disease remain unanswered: "Why does the host immune system fail to provide sterile immunity?" and "Why do only a proportion of infected individuals develop chronic pathology?" In this review, we describe the mechanisms proposed to explain the inability of the immune system to eradicate the parasite and the elements that allow the development of chronic heart disease. Moreover, we discuss the possibility that the inability of infected cardiomyocytes to sense intracellular T. cruzi contributes to parasite persistence in the heart and the development of chronic pathology.


Subject(s)
Chagas Disease/immunology , Animals , Chagas Disease/complications , Heart Diseases/etiology , Heart Diseases/immunology , Heart Diseases/parasitology , Humans , Myocytes, Cardiac/immunology , Myocytes, Cardiac/parasitology , Trypanosoma cruzi/immunology , Trypanosoma cruzi/pathogenicity
14.
PLoS Pathog ; 10(7): e1004188, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24991816

ABSTRACT

The purinergic P2X7 receptor (P2X7R) is a sensor of extracellular ATP, a damage-associated molecule that is released from necrotic cells and that induces pro-inflammatory cytokine production and cell death. To investigate whether the innate immune response to damage signals could contribute to the development of pulmonary necrotic lesions in severe forms of tuberculosis, disease progression was examined in C57BL/6 and P2X7R-/- mice that were intratracheally infected with highly virulent mycobacterial strains (Mycobacterium tuberculosis strain 1471 of the Beijing genotype family and Mycobacterium bovis strain MP287/03). The low-dose infection of C57BL/6 mice with bacteria of these strains caused the rapid development of extensive granulomatous pneumonia with necrotic areas, intense bacillus dissemination and anticipated animal death. In contrast, in P2X7R-/- mice, the lung pathology presented with moderate infiltrates of mononuclear leukocytes without visible signs of necrosis; the disease attenuation was accompanied by a delay in mortality. In vitro, the hypervirulent mycobacteria grew rapidly inside macrophages and induced death by a P2X7R-dependent mechanism that facilitated the release of bacilli. Furthermore, these bacteria were resistant to the protective mechanisms elicited in macrophages following extracellular ATP stimulation. Based on this study, we propose that the rapid intracellular growth of hypervirulent mycobacteria results in massive macrophage damage. The ATP released by damaged cells engages P2X7R and accelerates the necrotic death of infected macrophages and the release of bacilli. This vicious cycle exacerbates pneumonia and lung necrosis by promoting widespread cell destruction and bacillus dissemination. These findings suggest the use of drugs that have been designed to inhibit the P2X7R as a new therapeutic approach to treat the aggressive forms of tuberculosis.


Subject(s)
Macrophages , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Receptors, Purinergic P2X7 , Tuberculosis, Pulmonary , Adenosine Triphosphate/immunology , Animals , Humans , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Mycobacterium bovis/immunology , Mycobacterium bovis/pathogenicity , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/immunology , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology
15.
PLoS Negl Trop Dis ; 7(10): e2469, 2013.
Article in English | MEDLINE | ID: mdl-24098823

ABSTRACT

Trypanosoma cruzi (T. cruzi) is an intracellular protozoan parasite and the etiological agent of Chagas disease, a chronic infectious illness that affects millions of people worldwide. Although the role of TLR and Nod1 in the control of T. cruzi infection is well-established, the involvement of inflammasomes remains to be elucidated. Herein, we demonstrate for the first time that T. cruzi infection induces IL-1ß production in an NLRP3- and caspase-1-dependent manner. Cathepsin B appears to be required for NLRP3 activation in response to infection with T. cruzi, as pharmacological inhibition of cathepsin B abrogates IL-1ß secretion. NLRP3(-/-) and caspase1(-/-) mice exhibited high numbers of T. cruzi parasites, with a magnitude of peak parasitemia comparable to MyD88(-/-) and iNOS(-/-) mice (which are susceptible models for T. cruzi infection), indicating the involvement of NLRP3 inflammasome in the control of the acute phase of T. cruzi infection. Although the inflammatory cytokines IL-6 and IFN-γ were found in spleen cells from NLRP3(-/-) and caspase1(-/-) mice infected with T. cruzi, these mice exhibited severe defects in nitric oxide (NO) production and an impairment in macrophage-mediated parasite killing. Interestingly, neutralization of IL-1ß and IL-18, and IL-1R genetic deficiency demonstrate that these cytokines have a minor effect on NO secretion and the capacity of macrophages to control T. cruzi infection. In contrast, inhibition of caspase-1 with z-YVAD-fmk abrogated NO production by WT and MyD88(-/-) macrophages and rendered them as susceptible to T. cruzi infection as NLRP3(-/-) and caspase-1(-/-) macrophages. Taken together, our results demonstrate a role for the NLRP3 inflammasome in the control of T. cruzi infection and identify NLRP3-mediated, caspase-1-dependent and IL-1R-independent NO production as a novel effector mechanism for these innate receptors.


Subject(s)
Carrier Proteins/metabolism , Caspase 1/metabolism , Chagas Disease/immunology , Host-Pathogen Interactions , Interleukin-1beta/biosynthesis , Nitric Oxide/metabolism , Trypanosoma cruzi/immunology , Animals , Chagas Disease/parasitology , Disease Models, Animal , Female , Gene Expression Regulation , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein
16.
Proc Natl Acad Sci U S A ; 110(35): E3321-30, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23942123

ABSTRACT

NAIP5/NLRC4 (neuronal apoptosis inhibitory protein 5/nucleotide oligomerization domain-like receptor family, caspase activation recruitment domain domain-containing 4) inflammasome activation by cytosolic flagellin results in caspase-1-mediated processing and secretion of IL-1ß/IL-18 and pyroptosis, an inflammatory cell death pathway. Here, we found that although NLRC4, ASC, and caspase-1 are required for IL-1ß secretion in response to cytosolic flagellin, cell death, nevertheless, occurs in the absence of these molecules. Cytosolic flagellin-induced inflammasome-independent cell death is accompanied by IL-1α secretion and is temporally correlated with the restriction of Salmonella Typhimurium infection. Despite displaying some apoptotic features, this peculiar form of cell death do not require caspase activation but is regulated by a lysosomal pathway, in which cathepsin B and cathepsin D play redundant roles. Moreover, cathepsin B contributes to NAIP5/NLRC4 inflammasome-induced pyroptosis and IL-1α and IL-1ß production in response to cytosolic flagellin. Together, our data describe a pathway induced by cytosolic flagellin that induces a peculiar form of cell death and regulates inflammasome-mediated effector mechanisms of macrophages.


Subject(s)
Cytosol/metabolism , Flagellin/metabolism , Inflammasomes/metabolism , Lysosomes/metabolism , Macrophages/immunology , Animals , Apoptosis , Cells, Cultured , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Toll-Like Receptor 5/genetics
17.
Nat Med ; 19(7): 909-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23749230

ABSTRACT

Parasites of the Leishmania genus are the causative agents of leishmaniasis in humans, a disease that affects more than 12 million people worldwide. These parasites replicate intracellularly in macrophages, and the primary mechanisms underlying host resistance involve the production of nitric oxide (NO). In this study we show that the Nlrp3 inflammasome is activated in response to Leishmania infection and is important for the restriction of parasite replication both in macrophages and in vivo as demonstrated through the infection of inflammasome-deficient mice with Leishmania amazonensis, Leishmania braziliensis and Leishmania infantum chagasi. Inflammasome-driven interleukin-1ß (IL-1ß) production facilitated host resistance to infection, as signaling through IL-1 receptor (IL-1R) and MyD88 was necessary and sufficient to trigger inducible nitric oxide synthase (NOS2)-mediated production of NO. In this manuscript we identify a major signaling platform for host resistance to Leishmania spp. infection and describe the molecular mechanisms underlying Leishmania-induced NO production.


Subject(s)
Disease Resistance/drug effects , Inflammasomes/immunology , Interleukin-1beta/metabolism , Leishmania , Nitric Oxide/immunology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Disease Resistance/genetics , Disease Resistance/immunology , Female , Leishmaniasis/genetics , Leishmaniasis/immunology , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nitric Oxide Synthase Type II/genetics
18.
Methods ; 61(2): 110-6, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23454287

ABSTRACT

Pyroptosis is a molecularly controlled form of cell death that exhibits some features of apoptosis as well of necrosis. Pyroptosis is induced by inflammasome-activated caspase-1 or caspase-11 (caspase-4 in humans), as a result of distinct pathogenic or damage stimuli. Although pyroptosis displays some morphological and biochemical features of apoptosis, it has an inflammatory outcome due to the loss of plasma membrane integrity and the consequent release of intracellular contents, reminiscent to necrosis. Here, we use cytosolic delivery of purified flagellin as an experimental tool to trigger pyroptosis and describe potential methods to study this form of cell death. Finally, we discuss the advantages and limitations of these methods.


Subject(s)
Apoptosis/genetics , Cytosol/metabolism , Flagellin/metabolism , Inflammasomes/metabolism , Macrophages, Peritoneal/metabolism , Animals , Bacillus subtilis/chemistry , Caspase 1/deficiency , Caspase 1/genetics , Cytosol/ultrastructure , Enzyme Activation , Fatty Acids, Monounsaturated/chemistry , Flagellin/pharmacology , Humans , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/genetics , Necrosis/pathology , Protein Transport , Quaternary Ammonium Compounds/chemistry , Signal Transduction
19.
J Immunol ; 190(7): 3629-38, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460746

ABSTRACT

Pathogens are detected by innate immune receptors that, upon activation, orchestrate an appropriate immune response. Recent studies revealed the intracellular signaling cascades involved in the TLR-initiated immune response to Brucella abortus infection. However, no report has elucidated the role of inflammasome receptors in Brucella recognition. Therefore, we decided to investigate the function of NLRC4, NLRP3, and AIM2 in sensing Brucella. In this study, we showed that NLRC4 is not required to induce caspase-1 activation and further secretion of IL-1ß by B. abortus in macrophages. In contrast, we determined that AIM2, which senses Brucella DNA, and NLRP3 are partially required for caspase-1 activation and IL-1ß secretion. Additionally, mitochondrial reactive oxygen species induced by Brucella were implicated in IL-1ß production. Furthermore, AIM2, NLRP3, ASC, and caspase-1 knockout mice were more susceptible to B. abortus infection than were wild-type animals, suggesting that multiple ASC-dependent inflammasomes contribute to host protection against infection. This protective effect is due to the inflammatory response caused by IL-1ß and IL-18 rather than pyroptosis, because we observed augmented bacterial burden in IL-1R and IL-18 knockout mice. Finally, we determined that bacterial type IV secretion system VirB and live, but not heat-killed, Brucella are required for full inflammasome activation in macrophages during infection. Taken together, our results indicate that Brucella is sensed by ASC inflammasomes that collectively orchestrate a robust caspase-1 activation and proinflammatory response.


Subject(s)
Bacterial Secretion Systems , Brucella abortus/immunology , Brucella abortus/metabolism , Brucellosis/immunology , Brucellosis/metabolism , Caspase 1/metabolism , Cytoskeletal Proteins/metabolism , Inflammasomes , Animals , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , Calcium-Binding Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/genetics , DNA, Bacterial/metabolism , DNA-Binding Proteins , Enzyme Activation , Genetic Predisposition to Disease , Granuloma/immunology , Granuloma/metabolism , Granuloma/microbiology , Immunity, Innate , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Liver/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Reactive Oxygen Species/metabolism , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism
20.
PLoS Pathog ; 8(5): e1002699, 2012.
Article in English | MEDLINE | ID: mdl-22615561

ABSTRACT

MHC class Ia-restricted CD8(+) T cells are important mediators of the adaptive immune response against infections caused by intracellular microorganisms. Whereas antigen-specific effector CD8(+) T cells can clear infection caused by intracellular pathogens, in some circumstances, the immune response is suboptimal and the microorganisms survive, causing host death or chronic infection. Here, we explored the cellular and molecular mechanisms that could explain why CD8(+) T cell-mediated immunity during infection with the human protozoan parasite Trypanosoma cruzi is not optimal. For that purpose, we compared the CD8(+) T-cell mediated immune responses in mice infected with T. cruzi or vaccinated with a recombinant adenovirus expressing an immunodominant parasite antigen. Several functional and phenotypic characteristics of specific CD8(+) T cells overlapped. Among few exceptions was an accelerated expansion of the immune response in adenoviral vaccinated mice when compared to infected ones. Also, there was an upregulated expression of the apoptotic-signaling receptor CD95 on the surface of specific T cells from infected mice, which was not observed in the case of adenoviral-vaccinated mice. Most importantly, adenoviral vaccine provided at the time of infection significantly reduced the upregulation of CD95 expression and the proapoptotic phenotype of pathogen-specific CD8(+) cells expanded during infection. In parallel, infected adenovirus-vaccinated mice had a stronger CD8 T-cell mediated immune response and survived an otherwise lethal infection. We concluded that a suboptimal CD8(+) T-cell response is associated with an upregulation of CD95 expression and a proapoptotic phenotype. Both can be blocked by adenoviral vaccination.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neuraminidase/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , fas Receptor/biosynthesis , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Apoptosis , Chagas Disease/immunology , Chagas Disease/prevention & control , Interferon-gamma/biosynthesis , Lysosomal-Associated Membrane Protein 1/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Trypanosoma cruzi/pathogenicity , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...