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










Publication year range
1.
Infect Immun ; 90(9): e0032422, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35993771

ABSTRACT

The protozoan parasite Leishmania (L.) amazonensis infects and replicates inside host macrophages due to subversion of the innate host cell response. In the present study, we demonstrate that TLR3 is required for the intracellular growth of L. (L.) amazonensis. We observed restricted intracellular infection of TLR3-/- mouse macrophages, reduced levels of IFN1ß and IL-10, and increased levels of IL-12 upon L. (L.) amazonensis infection, compared with their wild-type counterparts. Accordingly, in vivo infection of TLR3-/- mice with L. (L.) amazonensis displayed a significant reduction in lesion size. Leishmania (L.) amazonensis infection induced TLR3 proteolytic cleavage, which is a process required for TLR3 signaling. The chemical inhibition of TLR3 cleavage or infection by CPB-deficient mutant L. (L.) mexicana resulted in reduced parasite load and restricted the expression of IFN1ß and IL-10. Furthermore, we show that the dsRNA sensor molecule PKR (dsRNA-activated protein kinase) cooperates with TLR3 signaling to potentiate the expression of IL-10 and IFN1ß and parasite survival. Altogether, our results show that TLR3 signaling is engaged during L. (L.) amazonensis infection and this component of innate immunity modulates the host cell response.


Subject(s)
Leishmania mexicana , Leishmaniasis , Parasites , Toll-Like Receptor 3 , Animals , Interleukin-10/metabolism , Interleukin-12/metabolism , Leishmania mexicana/metabolism , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Mice , Parasites/metabolism , Protein Kinases/metabolism , Toll-Like Receptor 3/metabolism
2.
Nat Commun ; 12(1): 3221, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050145

ABSTRACT

Lysine methylation on histone tails impacts genome regulation and cell fate determination in many developmental processes. Apicomplexa intracellular parasites cause major diseases and they have developed complex life cycles with fine-tuned differentiation events. Yet, apicomplexa genomes have few transcription factors and little is known about their epigenetic control systems. Tick-borne Theileria apicomplexa species have relatively small, compact genomes and a remarkable ability to transform leucocytes in their bovine hosts. Here we report enriched H3 lysine 18 monomethylation (H3K18me1) on the gene bodies of repressed genes in Theileria macroschizonts. Differentiation to merozoites (merogony) leads to decreased H3K18me1 in parasite nuclei. Pharmacological manipulation of H3K18 acetylation or methylation impacted parasite differentiation and expression of stage-specific genes. Finally, we identify a parasite SET-domain methyltransferase (TaSETup1) that can methylate H3K18 and represses gene expression. Thus, H3K18me1 emerges as an important epigenetic mark which controls gene expression and stage differentiation in Theileria parasites.


Subject(s)
Epigenetic Repression/physiology , Gene Expression Regulation, Developmental/physiology , Histones/metabolism , Life Cycle Stages/genetics , Theileria/growth & development , Acetylation/drug effects , Animals , Cattle , Cell Line , Chickens , Chromatin Immunoprecipitation Sequencing , Epigenetic Repression/drug effects , Gene Expression Regulation, Developmental/drug effects , HEK293 Cells , Humans , Insect Proteins/metabolism , Life Cycle Stages/drug effects , Lysine/metabolism , Methylation/drug effects , Methyltransferases/genetics , Methyltransferases/isolation & purification , Methyltransferases/metabolism , Mutagenesis, Site-Directed , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , RNA-Seq , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Theileria/genetics , Theileriasis/drug therapy , Theileriasis/parasitology , Tranylcypromine/pharmacology , Tranylcypromine/therapeutic use
3.
FASEB J ; 33(10): 10794-10807, 2019 10.
Article in English | MEDLINE | ID: mdl-31284755

ABSTRACT

Visceral leishmaniasis is a deadly illness caused by Leishmania donovani that provokes liver and spleen inflammation and tissue destruction. In cutaneous leishmaniasis, the protein of L. major, named inhibitor of serine peptidases (ISP) 2, inactivates neutrophil elastase (NE) present at the macrophage surface, resulting in blockade of TLR4 activation, prevention of TNF-α and IFN-ß production, and parasite survival. We report poor intracellular growth of L. donovani in macrophages from knockout mice for NE (ela-/-), TLR4, or TLR2. NE and TLR4 colocalized with the parasite in the parasitophorous vacuole. Parasite load in the liver and spleen of ela-/- mice were reduced and accompanied by increased NO and decreased TGF-ß production. Expression of ISP2 was not detected in L. donovani, and a transgenic line constitutively expressing ISP2, displayed poor intracellular growth in macrophages and decreased burden in mice. Infected ela-/- macrophages displayed significantly lower IFN-ß mRNA than background mice macrophages, and the intracellular growth was fully restored by exogenous IFN-ß. We propose that L. donovani utilizes the host NE-TLR machinery to induce IFN-ß necessary for parasite survival and growth during early infection. Low or absent expression of parasite ISP2 in L. donovani is necessary to preserve the activation of the NE-TLR pathway.-Dias, B. T., Dias-Teixeira, K. L., Godinho, J. P., Faria, M. S., Calegari-Silva, T., Mukhtar, M. M., Lopes, U. G., Mottram, J. C., Lima, A. P. C. A. Neutrophil elastase promotes Leishmania donovani infection via interferon-ß.


Subject(s)
Interferon-beta/metabolism , Leishmania donovani/pathogenicity , Leishmaniasis, Visceral/etiology , Leukocyte Elastase/metabolism , Animals , Animals, Genetically Modified , Leishmania donovani/genetics , Leishmania donovani/physiology , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Leukocyte Elastase/deficiency , Leukocyte Elastase/genetics , Macrophages/metabolism , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protozoan Proteins/genetics , Protozoan Proteins/physiology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
4.
PLoS Negl Trop Dis ; 13(6): e0007500, 2019 06.
Article in English | MEDLINE | ID: mdl-31216268

ABSTRACT

BACKGROUND: Leishmania parasites are transmitted to vertebrate hosts by phlebotomine sandflies and, in humans, may cause tegumentary or visceral leishmaniasis. The role of PKR (dsRNA activated kinase) and Toll-like receptor 3 (TLR3) activation in the control of Leishmania infection highlights the importance of the engagement of RNA sensors, which are usually involved in the antiviral cell response, in the fate of parasitism by Leishmania. We tested the hypothesis that Phlebovirus, a subgroup of the Bunyaviridae, transmitted by sandflies, would interfere with Leishmania infection. METHODOLOGY/PRINCIPAL FINDINGS: We tested two Phlebovirus isolates, Icoaraci and Pacui, from the rodents Nectomys sp. and Oryzomys sp., respectively, both natural sylvatic reservoir of Leishmania (Leishmania) amazonensis from the Amazon region. Phlebovirus coinfection with L. (L.) amazonensis in murine macrophages led to increased intracellular growth of L. (L.) amazonensis. Further studies with Icoaraci coinfection revealed the requirement of the PKR/IFN1 axis on the exacerbation of the parasite infection. L. (L.) amazonensis and Phlebovirus coinfection potentiated PKR activation and synergistically induced the expression of IFNß and IL-10. Importantly, in vivo coinfection of C57BL/6 mice corroborated the in vitro data. The exacerbation effect of RNA virus on parasite infection may be specific because coinfection with dengue virus (DENV2) exerted the opposite effect on parasite load. CONCLUSIONS: Altogether, our data suggest that coinfections with specific RNA viruses shared by vectors or reservoirs of Leishmania may enhance and sustain the activation of host cellular RNA sensors, resulting in aggravation of the parasite infection. The present work highlights new perspectives for the investigation of antiviral pathways as important modulators of protozoan infections.


Subject(s)
Bunyaviridae Infections/complications , Coinfection/immunology , Disease Susceptibility , Interferon-beta/metabolism , Interleukin-10/metabolism , Leishmaniasis/immunology , eIF-2 Kinase/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Leishmania/immunology , Mice, Inbred C57BL , Models, Theoretical , Phlebovirus/immunology
5.
Eur J Immunol ; 48(7): 1188-1198, 2018 07.
Article in English | MEDLINE | ID: mdl-29645094

ABSTRACT

The induced expression of nitric oxide synthase (iNOS) controls the intracellular growth of Leishmania in infected macrophages. Histones deacetylases (HDACs) negatively regulate gene expression through the formation of complexes containing transcription factors such as NF-κB p50/50. Herein, we demonstrated the occupancy of p50/p50_HDAC1 to iNOS promoter associated with reduced levels of H3K9Ac. Remarkably, we found increased levels of HDAC1 in L. amazonensis-infected macrophages. HDAC1 upregulation was not found in L. major-infected macrophages. The parasite intracellular load was reduced in HDAC1 knocked-down macrophages, which presented increased nitric oxide levels. HDAC1 silencing led to the occupancy of CBP/p300 to iNOS promoter and the rise of H3K9Ac modification. Importantly, the immunostaining of skin samples from hiporeactive cutaneous leishmaniasis patients infected with L. amazonensis, revealed high levels of HDAC1. In brief, L. amazonensis induces HDAC1 in infected macrophages, which contribute to parasite survival and is associated to hiporeactive stage found in L. amazonensis infected patients.


Subject(s)
Histone Deacetylase 1/metabolism , Leishmania braziliensis/physiology , Leishmaniasis, Cutaneous/immunology , Macrophages/immunology , Nitric Oxide Synthase Type II/metabolism , Skin/pathology , Adolescent , Adult , Cells, Cultured , Child , Extinction, Biological , Female , Histone Deacetylase 1/genetics , Host-Parasite Interactions , Humans , Immune Evasion , Leishmaniasis, Cutaneous/genetics , Male , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Parasite Load , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Small Interfering/genetics , Sp1 Transcription Factor/metabolism , Young Adult
6.
Sci Rep ; 7(1): 17074, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213084

ABSTRACT

Leishmania parasites utilize adaptive evasion mechanisms in infected macrophages to overcome host defenses and proliferate. We report here that the PERK/eIF2α/ATF4 signaling branch of the integrated endoplasmic reticulum stress response (IERSR) is activated by Leishmania and this pathway is important for Leishmania amazonensis infection. Knocking down PERK or ATF4 expression or inhibiting PERK kinase activity diminished L. amazonensis infection. Knocking down ATF4 decreased NRF2 expression and its nuclear translocation, reduced HO-1 expression and increased nitric oxide production. Meanwhile, the increased expression of ATF4 and HO-1 mRNAs were observed in lesions derived from patients infected with the prevalent related species L.(V.) braziliensis. Our data demonstrates that Leishmania parasites activate the PERK/eIF2α/ATF-4 pathway in cultured macrophages and infected human tissue and that this pathway is important for parasite survival and progression of the infection.


Subject(s)
Activating Transcription Factor 4/metabolism , Eukaryotic Initiation Factor-2/metabolism , Leishmaniasis, Cutaneous/pathology , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Animals , Endoplasmic Reticulum Stress , HEK293 Cells , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Leishmania/pathogenicity , Leishmaniasis, Cutaneous/metabolism , Macrophages/cytology , Macrophages/metabolism , Macrophages/parasitology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Phosphorylation , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism
7.
Front Immunol ; 8: 1127, 2017.
Article in English | MEDLINE | ID: mdl-28959260

ABSTRACT

Leishmania parasites infect macrophages, causing a wide spectrum of human diseases, from cutaneous to visceral forms. In search of novel therapeutic targets, we performed comprehensive in vitro and ex vivo mapping of the signaling pathways upstream and downstream of antioxidant transcription factor [nuclear factor erythroid 2-related factor 2 (Nrf2)] in cutaneous leishmaniasis (CL), by combining functional assays in human and murine macrophages with a systems biology analysis of in situ (skin biopsies) CL patient samples. First, we show the PKR pathway controls the expression and activation of Nrf2 in Leishmania amazonensis infection in vitro. Nrf2 activation also required PI3K/Akt signaling and autophagy mechanisms. Nrf2- or PKR/Akt-deficient macrophages exhibited increased levels of ROS/RNS and reduced expression of Sod1 Nrf2-dependent gene and reduced parasite load. L. amazonensis counteracted the Nrf2 inhibitor Keap1 through the upregulation of p62 via PKR. This Nrf2/Keap1 observation was confirmed in situ in skin biopsies from Leishmania-infected patients. Next, we explored the ex vivo transcriptome in CL patients, as compared to healthy controls. We found the antioxidant response element/Nrf2 signaling pathway was significantly upregulated in CL, including downstream target p62. In silico enrichment analysis confirmed upstream signaling by interferon and PI3K/Akt, and validated our in vitro findings. Our integrated in vitro, ex vivo, and in silico approach establish Nrf2 as a central player in human cutaneous leishmaniasis and reveal Nrf2/PKR crosstalk and PI3K/Akt pathways as potential therapeutic targets.

8.
FASEB J ; 30(4): 1557-65, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26678450

ABSTRACT

Endoplasmic reticulum (ER) stress triggers the integrated ER-stress response (IERSR) that ensures cellular survival of ER stress and represents a primordial form of innate immunity. We investigated the role of IERSR duringLeishmania amazonensisinfection. Treatment of RAW 264.7 infected macrophages with the ER stress-inducing agent thapsigargin (TG; 1 µM) increasedL. amazonensisinfectivity in an IFN1-α receptor (IFNAR)-dependent manner. In Western blot assays, we showed thatL. amazonensisactivates the inositol-requiring enzyme (IRE1)/ X-box binding protein (XBP)-1-splicing arms of the IERSR in host cells. In chromatin immunoprecipitation (ChIP) assays, we showed an increased occupancy of enhancer and promoter sequences for theIfnbgene by XBP1 in infected RAW 264.7 cells. Knocking down XBP1 expression by transducing RAW 264.7 cells with the short hairpin XBP1 lentiviral vector significantly reduced the parasite proliferation associated with impaired translocation of phosphorylated IFN regulatory transcription factor (IRF)-3 to the nucleus and a decrease in IFN1-ß expression. Knocking down XBP1 expression also increased NO concentration, as determined by Griess reaction and reduced the expression of antioxidant genes, such as heme oxygenase (HO)-1, that protect parasites from oxidative stress. We conclude thatL. amazonensisactivation of XBP1 plays a critical role in infection by protecting the parasites from oxidative stress and increasing IFN1-ß expression.-Dias-Teixeira, K. L., Calegari-Silva, T. C., Dos Santos, G. R. R. M., Vitorino dos Santos, J., Lima, C., Medina, J. M., Aktas, B. H., Lopes, U. G. The integrated endoplasmic reticulum stress response inLeishmania amazonensismacrophage infection: the role of X-box binding protein 1 transcription factor.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Leishmania/physiology , Macrophages/metabolism , Macrophages/microbiology , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line , DNA-Binding Proteins/genetics , Gene Expression , HEK293 Cells , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Host-Parasite Interactions , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Macrophages/drug effects , Mice , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reactive Oxygen Species/metabolism , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Thapsigargin/pharmacology , Transcription Factors/genetics , X-Box Binding Protein 1
9.
Open Biol ; 5(9): 150118, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26400473

ABSTRACT

Leishmania amazonensis activates the NF-κB transcriptional repressor homodimer (p50/p50) and promotes nitric oxide synthase (iNOS) downregulation. We investigated the role of PI3K/Akt in p50/p50 NF-κB activation and the effect on iNOS expression in L. amazonensis infection. The increased occupancy of p50/p50 on the iNOS promoter of infected macrophages was observed and we demonstrated that both p50/p50 NF-κB induction and iNOS downregulation in infected macrophages depended on PI3K/Akt activation. Importantly, the intracellular growth of the parasite was also impaired during PI3K/Akt signalling inhibition and in macrophages knocked-down for Akt 1 expression. It was also observed that the increased nuclear levels of p50/p50 in L. amazonensis-infected macrophages were associated with reduced phosphorylation of 907 Ser p105, the precursor of p50. Corroborating these data, we demonstrated the increased levels of phospho-9 Ser GSK3ß in infected macrophages, which is associated with GSK3ß inhibition and, consequently, its inability to phosphorylate p105. Remarkably, we found that the levels of pPTEN 370 Ser, a negative regulator of PI3K, increased due to L. amazonensis infection. Our data support the notion that PI3K/Akt activity is sustained during the parasite infection, leading to NF-κB 105 phosphorylation and further processing to originate p50/p50 homodimers and the consequent downregulation of iNOS expression.


Subject(s)
Leishmania/physiology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Dimerization , Down-Regulation , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Leishmania/genetics , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Leishmaniasis/pathology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/chemistry , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/metabolism , Signal Transduction
10.
Immunobiology ; 220(4): 437-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466588

ABSTRACT

The protozoan parasite Leishmania infects and replicates in macrophages, causing a spectrum of diseases in the human host, varying from cutaneous to visceral clinical forms. It is known that cytokines modulate the immunological response against Leishmania and are relevant for infection resolution. Here, we report that Interleukin (IL)-27 increases Leishmania amazonensis replication in human and murine macrophages and that the blockage of the IL-10 receptor on the surface of infected cells abolished the IL-27-mediated enhancement of Leishmania growth. IL-27 induced the activation/phosphorylation of protein kinase R (PKR) in macrophages, and PKR blockage or PKR gene deletion abrogated the enhancement of the parasite growth driven by IL-27, as well as the L. amazonensis-induced macrophage production of IL-27. We also observed that L. amazonensis-induced expression of IL-27 depends on type I interferon signaling and the engagement of Toll-like receptor 2. Treatment of Leishmania-infected mice with IL-27 increased lesion size and parasite loads in the footpad and lymph nodes of infected animals, indicating that this cytokine exerts a local and a systemic effect on parasite growth and propagation. In conclusion, we show that IL-27 is a L. amazonensis-enhancing factor and that the PKR/IFN1 axis and IL-10 are critical mediators of this IL-27 induced effect.


Subject(s)
Interleukin-10/metabolism , Interleukin-27/metabolism , Leishmania mexicana , Leishmaniasis, Cutaneous/metabolism , Signal Transduction , eIF-2 Kinase/metabolism , Animals , Cell Line , Humans , Interferon Type I/metabolism , Interleukin-27/pharmacology , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/parasitology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/parasitology , Male , Mice , Mice, Knockout , Signal Transduction/drug effects , Toll-Like Receptor 2/metabolism , eIF-2 Kinase/genetics
11.
PLoS Negl Trop Dis ; 6(9): e1787, 2012.
Article in English | MEDLINE | ID: mdl-22970332

ABSTRACT

BACKGROUND: Chemotherapy for leishmaniasis, a disease caused by Leishmania parasites, is expensive and causes side effects. Furthermore, parasite resistance constitutes an increasing problem, and new drugs against this disease are needed. In this study, we examine the effect of the compound 8,10,18-trihydroxy-2,6-dolabelladiene (Dolabelladienetriol), on Leishmania growth in macrophages. The ability of this compound to modulate macrophage function is also described. METHODOLOGY/PRINCIPAL FINDINGS: Leishmania-infected macrophages were treated with Dolabelladienetriol, and parasite growth was measured using an infectivity index. Nitric oxide (NO), TNF-α and TGF-ß production were assayed in macrophages using specific assays. NF-kB nuclear translocation was analyzed by western blot. Dolabelladienetriol inhibited Leishmania in a dose-dependent manner; the IC(50) was 44 µM. Dolabelladienetriol diminished NO, TNF-α and TGF-ß production in uninfected and Leishmania-infected macrophages and reduced NF-kB nuclear translocation. Dolabelladienetriol inhibited Leishmania infection even when the parasite growth was exacerbated by either IL-10 or TGF-ß. In addition, Dolabelladienetriol inhibited Leishmania growth in HIV-1-co-infected human macrophages. CONCLUSION: Our results indicate that Dolabelladienetriol significantly inhibits Leishmania in macrophages even in the presence of factors that exacerbate parasite growth, such as IL-10, TGF-ß and HIV-1 co-infection. Our results suggest that Dolabelladienetriol is a promising candidate for future studies regarding treatment of leishmaniasis, associated or not with HIV-1 infection.


Subject(s)
Antiprotozoal Agents/pharmacology , Cell Extracts/pharmacology , Diterpenes/pharmacology , Leishmania/drug effects , Phaeophyceae/chemistry , Animals , Antiprotozoal Agents/isolation & purification , Cell Extracts/isolation & purification , Cells, Cultured , Diterpenes/isolation & purification , Humans , Inhibitory Concentration 50 , Leishmania/growth & development , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Nitric Oxide/metabolism , Parasitic Sensitivity Tests , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
PLoS One ; 7(7): e41772, 2012.
Article in English | MEDLINE | ID: mdl-22848596

ABSTRACT

ExoU, a Pseudomonas aeruginosa cytotoxin injected into host cytosol by type III secretion system, exhibits a potent proinflammatory activity that leads to a marked recruitment of neutrophils to infected tissues. To evaluate the mechanisms that account for neutrophil infiltration, we investigated the effect of ExoU on IL-8 secretion and NF-κB activation. We demonstrate that ExoU increases IL-8 mRNA and protein levels in P. aeruginosa-infected epithelial and endothelial cell lines. Also, ExoU induces the nuclear translocation of p65/p50 NF-κB transactivator heterodimer as well as NF-κB-dependent transcriptional activity. ChIP assays clearly revealed that ExoU promotes p65 binding to NF-κB site in IL-8 promoter and the treatment of cultures with the NF-κB inhibitor Bay 11-7082 led to a significant reduction in IL-8 mRNA levels and protein secretion induced by ExoU. These results were corroborated in a murine model of pneumonia that revealed a significant reduction in KC secretion and neutrophil infiltration in bronchoalveolar lavage when mice were treated with Bay 11-7082 before infection with an ExoU-producing strain. In conclusion, our data demonstrate that ExoU activates NF-κB, stimulating IL-8 expression and secretion during P. aeruginosa infection, and unveils a new mechanism triggered by this important virulence factor to interfere in host signaling pathways.


Subject(s)
Bacterial Proteins/metabolism , Interleukin-8/metabolism , NF-kappa B/metabolism , Pseudomonas aeruginosa/physiology , Animals , Bacterial Proteins/biosynthesis , Bronchoalveolar Lavage Fluid/microbiology , Capillaries/cytology , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Gene Expression Regulation , Interleukin-8/genetics , Mice , Neutrophil Infiltration , Pseudomonas aeruginosa/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory System/immunology , Respiratory System/microbiology
13.
FASEB J ; 25(12): 4162-73, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21846836

ABSTRACT

We investigated the type I interferon (IFN-1)/PKR axis in the outcome of the Leishmania (Leishmania) amazonensis infection, along with the underlying mechanisms that trigger and sustain this signaling pathway. Reporter assays of cell extracts from RAW-264.7 macrophages infected with L. (L.) amazonensis or HEK-293T cells cotransfected with TLR2 and PKR promoter constructions were employed. Primary macrophages of TLR2-knockout (KO) or IFNR-KO mice were infected, and the levels of PKR, IFN-1, and superoxide dismutase 1 (SOD1) transcript levels were investigated and compared. Immunohistochemical analysis of human biopsy lesions was evaluated for IFN-1 and PKR-positive cells. Leishmania infection increased the expression of PKR and IFN-ß on induction of PKR-promoter activity. The observed effects required the engagement of TLR2. TLR2-KO macrophages expressed low IFN-ß and PKR levels postinfection with a reduced parasite load. We also revealed the requirement of PKR signaling for Leishmania-induced IFN-1 expression, responsible for sustaining PKR expression and enhancing infection. Moreover, during infection, SOD1 transcripts increased and were also enhanced when IFN-1 was added to the cultures. Remarkably, SOD1 expression was abrogated in infected, dominant-negative PKR-expressing cells. Finally, lesions of patients with anergic diffuse cutaneous leishmaniasis exhibited higher levels of PKR/IFN-1-expressing cells compared to those with single cutaneous leishmaniasis. In summary, we demonstrated the mechanisms and relevance of the IFN-1/PKR axis in the Leishmania infection.


Subject(s)
Interferon Type I/metabolism , Leishmania mexicana , Leishmaniasis, Cutaneous/enzymology , Leishmaniasis, Cutaneous/immunology , Toll-Like Receptor 2/metabolism , eIF-2 Kinase/metabolism , Animals , Glycosphingolipids/immunology , Host-Parasite Interactions , Humans , Leishmania mexicana/immunology , Leishmania mexicana/pathogenicity , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Diffuse Cutaneous/enzymology , Leishmaniasis, Diffuse Cutaneous/genetics , Leishmaniasis, Diffuse Cutaneous/immunology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Promoter Regions, Genetic , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Transfection , eIF-2 Kinase/genetics
14.
FASEB J ; 24(2): 617-26, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19812373

ABSTRACT

The evolution of Leishmania infection depends on the balance between microbicidal and suppressor macrophage functions. Double-stranded RNA (dsRNA)-activated protein kinase R (PKR), a classic antiviral protein, is able to regulate a number of signaling pathways and macrophage functions. We investigated the possible role of PKR in the modulation of Leishmania infection. Our data demonstrated that Leishmania amazonensis infection led to PKR activation and increased PKR levels. Consistently, in macrophages from PKR knockout 129Sv/Ev mice and RAW-264.7 cells stably expressing a dominant-negative (DN) construct of PKR (DN-PKR), L. amazonensis infection was strongly reduced. The treatment of infected macrophages with the synthetic double-stranded RNA poly(I:C), a potent PKR inductor, increased L. amazonensis intracellular proliferation. This effect was reversed by 2-aminopurine (2-AP), a pharmacological inhibitor of PKR, as well as by the expression of DN-PKR. NO release induced by dsRNA treatment was inhibited by L. amazonensis through NF-kappaB modulation. PKR activation induced by dsRNA also resulted in IL-10 production, whose neutralization with specific antibody completely abrogated L. amazonensis proliferation. Our data demonstrated a new role of PKR in protozoan parasitic infection through IL-10 modulation.


Subject(s)
Leishmania/pathogenicity , Macrophages/parasitology , eIF-2 Kinase/metabolism , 2-Aminopurine/pharmacology , Animals , Enzyme Activation , Humans , Interleukin-10/metabolism , Leishmania/genetics , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Poly I-C/pharmacology , RNA, Double-Stranded/genetics
15.
Immunol Lett ; 127(1): 19-26, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-19712696

ABSTRACT

Host invasion by pathogens is frequently associated with the activation of nuclear factor kappaB (NF-kappaB), which modulates the expression of genes involved in the immunological response of the host. However, pathogens may also subvert these mechanisms to secure their survival. We describe the effect of Leishmania amazonensis infection on NF-kappaB transcriptional factor activation in macrophages and the subsequent reduction in inducible nitric oxide synthase (iNOS) expression. L. amazonensis promastigote infection activates the p50/p50 NF-kappaB complex, a classic transcriptional repressor. Interestingly, L. amazonensis promotes the change of the classical p65/p50 NF-kappaB dimer induced by LPS, leading to the p50/p50 NF-kappaB complex activation in macrophages stimulated with LPS. Moreover, this parasite promotes the reduction of p65 total levels in infected macrophages. All these effects contribute to the observation that this parasite is able to restrain the NF-kappaB-dependent transcriptional activity induced by LPS. Strikingly, L. amazonensis reduces the mRNA levels of the iNOS in addition to protein expression and the production of nitric oxide in LPS-stimulated macrophages. Accordingly, as revealed by reporter-gene assays, L. amazonensis-induced iNOS repression requires NF-kappaB sites in the iNOS promoter region. In summary, our results suggest that L. amazonensis has developed an adaptive strategy to escape from host defense by activating the NF-kappaB repressor complex p50/p50. The activation of this specific host transcriptional response negatively regulates the expression of iNOS, favoring the establishment and success of L. amazonensis infection.


Subject(s)
Leishmania/immunology , Leishmaniasis/immunology , Macrophages/metabolism , NF-kappa B p50 Subunit/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Cell Line , Enzyme Repression , Host-Pathogen Interactions , Humans , Leishmania/pathogenicity , Leishmaniasis/enzymology , Leishmaniasis/genetics , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , NF-kappa B p50 Subunit/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Transcriptional Activation
16.
Biochem Biophys Res Commun ; 335(1): 20-6, 2005 Sep 16.
Article in English | MEDLINE | ID: mdl-16055086

ABSTRACT

Mycobacterium leprae, the causative agent of leprosy, invades peripheral nerve Schwann cells, resulting in deformities associated with this disease. NF-kappaB is an important transcription factor involved in the regulation of host immune antimicrobial responses. We aimed in this work to investigate NF-kappaB signaling pathways in the human ST88-14 Schwannoma cell line infected with M. leprae. Gel shift and supershift assays indicate that two NF-kappaB dimers, p65/p50 and p50/p50, translocate to the nucleus in Schwann cells treated with lethally irradiated M. leprae. Consistent with p65/p50 and p50/p50 activation, we observed IkappaB-alpha degradation and reduction of p105 levels. The nuclear translocation of p50/p50 complex due to M. leprae treatment correlated with repression of NF-kappaB-driven transcription induced by TNF-alpha. Moreover, thalidomide inhibited p50 homodimer nuclear translocation induced by M. leprae and consequently rescues Schwann cells from NF-kappaB-dependent transcriptional repression. Here, we report for the first time that M. leprae induces NF-kappaB activation in Schwann cells and thalidomide is able to modulate this activation.


Subject(s)
Mycobacterium leprae/physiology , NF-kappa B/metabolism , Schwann Cells/metabolism , Schwann Cells/microbiology , Transcription, Genetic , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Down-Regulation , Humans , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/chemistry , Schwann Cells/drug effects , Thalidomide/pharmacology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
17.
J Immunol ; 174(10): 6314-21, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879131

ABSTRACT

During Trypanosoma cruzi infection, T cells up-regulate caspase-8 activity. To assess the role of caspase-8 in T cell-mediated immunity, we investigated the effects of caspase-8 inhibition on T cells in viral FLIP (v-FLIP) transgenic mice. Compared with wild-type controls, increased parasitemia was observed in v-FLIP mice infected with T. cruzi. There was a profound decrease in expansion of both CD4 and CD8 T cell subsets in the spleens of infected v-FLIP mice. We did not find differences in activation ratios of T cells from transgenic or wild-type infected mice. However, the numbers of memory/activated CD4 and CD8 T cells were markedly reduced in v-FLIP mice, possibly due to defective survival. We also found decreased production of IL-2 and increased secretion of type 2 cytokines, IL-4 and IL-10, which could enhance susceptibility to infection. Similar, but less pronounced, alterations were observed in mice treated with the caspase-8 inhibitor, zIETD. Furthermore, blockade of caspase-8 by zIETD in vitro mimicked the effects observed on T. cruzi infection in vivo, affecting the generation of activated/memory T cells and T cell cytokine production. Caspase-8 is also required for NF-kappaB signaling upon T cell activation. Blockade of caspase-8 by either v-FLIP expression or treatment with zIETD peptide decreased NF-kappaB responses to TCR:CD3 engagement in T cell cultures. These results suggest a critical role for caspase-8 in the establishment of T cell memory, cell signaling, and regulation of cytokine responses during protozoan infection.


Subject(s)
Caspases/physiology , Chagas Disease/immunology , Cytokines/biosynthesis , Th2 Cells/enzymology , Th2 Cells/immunology , Trypanosoma cruzi/immunology , Animals , Caspase 8 , Caspase Inhibitors , Caspases/biosynthesis , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Chagas Disease/enzymology , Chagas Disease/genetics , Cytokines/metabolism , Genetic Predisposition to Disease , Immunity, Cellular/genetics , Immunity, Innate/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/pharmacology , Th2 Cells/cytology , Th2 Cells/metabolism , Up-Regulation/genetics , Up-Regulation/immunology , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...