Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
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.
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
3.
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.

4.
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
5.
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
6.
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
7.
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
8.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...