Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 15(8): e1007923, 2019 08.
Article in English | MEDLINE | ID: mdl-31449558

ABSTRACT

IL-1ß is a potent pro-inflammatory cytokine that promotes immunity and host defense, and its dysregulation is associated with immune pathology. Toxoplasma gondii infection of myeloid cells triggers the production and release of IL-1ß; however, the mechanisms regulating this pathway, particularly in human immune cells, are incompletely understood. We have identified a novel pathway of T. gondii induction of IL-1ß via a Syk-CARD9-NF-κB signaling axis in primary human peripheral blood monocytes. Syk was rapidly phosphorylated during T. gondii infection of primary monocytes, and inhibiting Syk with the pharmacological inhibitors R406 or entospletinib, or genetic ablation of Syk in THP-1 cells, reduced IL-1ß release. Inhibition of Syk in primary cells or deletion of Syk in THP-1 cells decreased parasite-induced IL-1ß transcripts and the production of pro-IL-1ß. Furthermore, inhibition of PKCδ, CARD9/MALT-1 and IKK reduced p65 phosphorylation and pro-IL-1ß production in T. gondii-infected primary monocytes, and genetic knockout of PKCδ or CARD9 in THP-1 cells also reduced pro-IL-1ß protein levels and IL-1ß release during T. gondii infection, indicating that Syk functions upstream of this NF-κB-dependent signaling pathway for IL-1ß transcriptional activation. IL-1ß release from T. gondii-infected primary human monocytes required the NLRP3-caspase-1 inflammasome, but interestingly, was independent of gasdermin D (GSDMD) cleavage and pyroptosis. Moreover, GSDMD knockout THP-1 cells released comparable amounts of IL-1ß to wild-type THP-1 cells after T. gondii infection. Taken together, our data indicate that T. gondii induces a Syk-CARD9/MALT-1-NF-κB signaling pathway and activation of the NLRP3 inflammasome for the release of IL-1ß in a cell death- and GSDMD-independent manner. This research expands our understanding of the molecular basis for human innate immune regulation of inflammation and host defense during parasite infection.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , Phosphate-Binding Proteins/metabolism , Syk Kinase/metabolism , Toxoplasmosis/metabolism , CARD Signaling Adaptor Proteins/genetics , Cells, Cultured , Humans , Inflammasomes , Intracellular Signaling Peptides and Proteins/genetics , Monocytes/immunology , Monocytes/microbiology , NF-kappa B/genetics , Phosphate-Binding Proteins/genetics , Signal Transduction , Syk Kinase/genetics , Toxoplasma/physiology , Toxoplasmosis/immunology , Toxoplasmosis/microbiology
2.
mBio ; 9(1)2018 02 13.
Article in English | MEDLINE | ID: mdl-29440572

ABSTRACT

Neutrophils are a major player in host immunity to infection; however, the mechanisms by which human neutrophils respond to the intracellular protozoan parasite Toxoplasma gondii are still poorly understood. In the current study, we found that, whereas primary human monocytes produced interleukin-1beta (IL-1ß) in response to T. gondii infection, human neutrophils from the same blood donors did not. Moreover, T. gondii inhibited lipopolysaccharide (LPS)-induced IL-1ß synthesis in human peripheral blood neutrophils. IL-1ß suppression required active parasite invasion, since heat-killed or mycalolide B-treated parasites did not inhibit IL-1ß release. By investigating the mechanisms involved in this process, we found that T. gondii infection of neutrophils treated with LPS resulted in reduced transcript levels of IL-1ß and NLRP3 and reduced protein levels of pro-IL-1ß, mature IL-1ß, and the inflammasome sensor NLRP3. In T. gondii-infected neutrophils stimulated with LPS, the levels of MyD88, TRAF6, IKKα, IKKß, and phosphorylated IKKα/ß were not affected. However, LPS-induced IκBα degradation and p65 phosphorylation were reduced in T. gondii-infected neutrophils, and degradation of IκBα was reversed by treatment with the proteasome inhibitor MG-132. Finally, we observed that T. gondii inhibited the cleavage and activity of caspase-1 in human neutrophils. These results indicate that T. gondii suppression of IL-1ß involves a two-pronged strategy whereby T. gondii inhibits both NF-κB signaling and activation of the NLRP3 inflammasome. These findings represent a novel mechanism of T. gondii evasion of human neutrophil-mediated host defense by targeting the production of IL-1ß.IMPORTANCEToxoplasma gondii is an obligate intracellular parasite that infects approximately one-third of humans worldwide and can invade virtually any nucleated cell in the human body. Although it is well documented that neutrophils infiltrate the site of acute T. gondii infection, there is limited understanding of how human neutrophils respond to T. gondii Neutrophils control infectious pathogens by a variety of mechanisms, including the release of the cytokine IL-1ß, a major driver of inflammation during infection. This study reveals that T. gondii is able to inhibit IL-1ß production in human neutrophils by impairing the activation of the NF-κB signaling pathway and by inhibiting the inflammasome, the protein complex responsible for IL-1ß maturation. This two-pronged strategy of targeting the IL-1ß pathway may facilitate the survival and spread of T. gondii during acute infection.


Subject(s)
Immune Evasion , Neutrophils/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Cells, Cultured , Healthy Volunteers , Humans , Immunologic Factors/metabolism , Interleukin-1beta/metabolism , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
3.
J Immunol ; 199(8): 2855-2864, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28904126

ABSTRACT

IL-1ß is produced by myeloid cells and acts as a critical mediator of host defense during infection and injury. We found that the intracellular protozoan parasite Toxoplasma gondii induced an early IL-1ß response (within 4 h) in primary human peripheral blood monocytes isolated from healthy donors. This process involved upregulation of IL-1ß, IL-1RN (IL-1R antagonist), and NLRP3 transcripts, de novo protein synthesis, and the release of pro- and mature IL-1ß from infected primary monocytes. The released pro-IL-1ß was cleavable to mature bioactive IL-1ß in the extracellular space by the protease caspase-1. Treatment of primary monocytes with the NLRP3 inhibitor MCC950 or with extracellular potassium significantly reduced IL-1ß cleavage and release in response to T. gondii infection, without affecting the release of TNF-α, and indicated a role for the inflammasome sensor NLRP3 and for potassium efflux in T. gondii-induced IL-1ß production. Interestingly, T. gondii infection did not induce an IL-1ß response in primary human macrophages derived from the same blood donors as the monocytes. Consistent with this finding, NLRP3 was downregulated during the differentiation of monocytes to macrophages and was not induced in macrophages during T. gondii infection. To our knowledge, these findings are the first to identify NLRP3 as an inflammasome sensor for T. gondii in primary human peripheral blood cells and to define an upstream regulator of its activation through the release of intracellular potassium.


Subject(s)
Inflammasomes/metabolism , Interleukin-1beta/metabolism , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium/metabolism , Toxoplasma/immunology , Toxoplasmosis/immunology , Cell Differentiation , Cells, Cultured , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Indenes , Macrophages/immunology , Monocytes/parasitology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Primary Cell Culture , Proteolysis/drug effects , Sulfonamides , Sulfones/pharmacology
4.
Infect Immun ; 82(10): 4047-55, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25024369

ABSTRACT

Toxoplasma gondii is an obligate intracellular parasite that can cause severe neurological disease in infected humans. CD40 is a receptor on macrophages that plays a critical role in controlling T. gondii infection. We examined the regulation of CD40 on the surface of T. gondii-infected bone marrow-derived macrophages (BMdMs). T. gondii induced CD40 expression both at the transcript level and on the cell surface, and interestingly, the effect was parasite strain specific: CD40 levels were dramatically increased in type II T. gondii-infected BMdMs compared to type I- or type III-infected cells. Type II induction of CD40 was specific to cells harboring intracellular parasites and detectable as early as 6 h postinfection (hpi) at the transcript level. CD40 protein expression peaked at 18 hpi. Using forward genetics with progeny from a type II × type III cross, we found that CD40 induction mapped to a region of chromosome X that included the gene encoding the dense granule protein 15 (GRA15). Using type I parasites stably expressing the type II allele of GRA15 (GRA15II), we found that type I GRA15II parasites induced the expression of CD40 on infected cells in an NF-κB-dependent manner. In addition, stable expression of hemagglutinin-tagged GRA15II in THP-1 cells resulted in CD40 upregulation in the absence of infection. Since CD40 signaling contributes to interleukin-12 (IL-12) production, we examined IL-12 from infected macrophages and found that CD40L engagement of CD40 amplified the IL-12 response in type II-infected cells. These data indicate that GRA15II induction of CD40 promotes parasite immunity through the production of IL-12.


Subject(s)
CD40 Antigens/biosynthesis , CD40 Antigens/immunology , Interleukin-12/immunology , Macrophages/immunology , Macrophages/parasitology , Protozoan Proteins/immunology , Toxoplasma/immunology , Animals , Antigens, Protozoan/immunology , Cells, Cultured , Humans
5.
mBio ; 4(4)2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23839215

ABSTRACT

UNLABELLED: Interleukin-1ß (IL-1ß) functions as a key regulator of inflammation and innate immunity. The protozoan parasite Toxoplasma gondii actively infects human blood monocytes and induces the production of IL-1ß; however, the host and parasite factors that mediate IL-1ß production during T. gondii infection are poorly understood. We report that T. gondii induces IL-1ß transcript, processing/cleavage, and release from infected primary human monocytes and THP-1 cells. Treating monocytes with the caspase-1 inhibitor Ac-YVAD-CMK reduced IL-1ß release, suggesting a role for the inflammasome in T. gondii-induced IL-1ß production. This was confirmed by performing short hairpin RNA (shRNA) knockdown of caspase-1 and of the inflammasome adaptor protein ASC. IL-1ß induction required active parasite invasion of monocytes, since heat-killed or mycalolide B-treated parasites did not induce IL-1ß. Among the type I, II, and III strains of T. gondii, the type II strain induced substantially more IL-1ß mRNA and protein release than did the type I and III strains. Since IL-1ß transcript is known to be induced downstream of NF-κB signaling, we investigated a role for the GRA15 protein, which induces sustained NF-κB signaling in a parasite strain-specific manner. By infecting human monocytes with a GRA15-knockout type II strain and a type I strain stably expressing type II GRA15, we determined that GRA15 is responsible for IL-1ß induction during T. gondii infection of human monocytes. This research defines a pathway driving human innate immunity by describing a role for the classical inflammasome components caspase-1 and ASC and the parasite GRA15 protein in T. gondii-induced IL-1ß production. IMPORTANCE: Monocytes are immune cells that protect against infection by increasing inflammation and antimicrobial activities in the body. Upon infection with the parasitic pathogen Toxoplasma gondii, human monocytes release interleukin-1ß (IL-1ß), a "master regulator" of inflammation, which amplifies immune responses. Although inflammatory responses are critical for host defense against infection, excessive inflammation can result in tissue damage and pathology. This delicate balance underscores the importance of understanding the mechanisms that regulate IL-1ß during infection. We have investigated the molecular pathway by which T. gondii induces the synthesis and release of IL-1ß in human monocytes. We found that specific proteins in the parasite and the host cell coordinate to induce IL-1ß production. This research is significant because it contributes to a greater understanding of human innate immunity to infection and IL-1ß regulation, thereby enhancing our potential to modulate inflammation in the body.


Subject(s)
Antigens, Protozoan/immunology , Caspase 1/immunology , Cytoskeletal Proteins/immunology , Immunity, Innate , Interleukin-1beta/immunology , Monocytes/immunology , Toxoplasma/immunology , Antigens, Protozoan/genetics , CARD Signaling Adaptor Proteins , Cells, Cultured , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Monocytes/parasitology , Toxoplasma/genetics
6.
J Neuroimmunol ; 254(1-2): 110-6, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23102608

ABSTRACT

Epstein-Barr virus-induced gene 3 (EBI3) associates with p28 and p35 to form the immunomodulatory cytokines IL-27 and IL-35, respectively. Infection of EBI3-/- mice with the neuroadapted JHM strain of mouse hepatitis virus (JHMV) resulted in increased mortality that was not associated with impaired ability to control viral replication but enhanced T cell and macrophage infiltration into the CNS. IFN-γ secretion from virus-specific CD4+ and CD8+ T cells isolated from infected EBI3-/- mice was augmented while IL-10 expression muted in comparison to infected WT mice. These data demonstrate a regulatory role for EBI3-associated cytokines in controlling host responses following CNS viral infection.


Subject(s)
Coronavirus Infections/complications , Encephalomyelitis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, Cytokine/metabolism , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , CD8 Antigens/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis/etiology , Encephalomyelitis/immunology , Encephalomyelitis/mortality , Encephalomyelitis/pathology , Flow Cytometry , Gene Expression Regulation, Viral/genetics , Glial Fibrillary Acidic Protein/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , RNA, Messenger/metabolism , Receptors, Cytokine/deficiency , T-Lymphocytes/virology , Time Factors
7.
Blood ; 114(2): 404-14, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19398721

ABSTRACT

MicroRNAs (miRNAs, miRs) modulate a multitude of cellular events. Here, we identify functional miRNA-protein networks that regulate human monocyte-derived dendritic cell (MDDC) differentiation. miRNA profiling revealed stage-specific differential expression of 20 miRNAs during days 1, 3, and 5 of MDDC differentiation. To identify and prioritize miRNA-protein networks for functional validation, we developed a target ranking algorithm that incorporates many features of miRNA regulatory networks. This system prioritized miR-21, miR-34a, and their cognate targets WNT1 and JAG1 for functional validation. Inhibition of both miR-21 and miR-34a stalled MDDC differentiation, as quantified by DC-SIGN/CD14 expression ratios, showing cooperative involvement of these miRNAs in MDDC differentiation. We confirmed that the 3' untranslated regions of WNT1 and JAG1 were functional targets of these miRNAs and provide evidence that these targets were translationally suppressed. Significantly, exogenously added Wnt-1 and Jagged-1 also stalled MDDC differentiation, suggesting that miRNA-mediated inhibition of endogenous WNT1 and JAG1 expression was important for proper MDDC differentiation. Finally, inhibition of miR-21 and miR-34a, or addition of Wnt-1 and Jagged-1, led to a decrease in endocytic capacity, a key function of immature DCs. Thus, our novel approach identified and validated some miRNA-protein networks involved in phenotypic and functional MDDC differentiation.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , MicroRNAs/genetics , Wnt1 Protein/metabolism , Calcium-Binding Proteins/genetics , Cell Line , Computational Biology , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Serrate-Jagged Proteins , Wnt1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...