Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Microb Pathog ; 173(Pt A): 105881, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36379373

ABSTRACT

Cardiomyopathy is the most serious complication of chronic Chagas disease, caused by infection with the protozoan Trypanosoma cruzi. Exacerbated inflammation of the myocardium constitutes a major pathologic component of the disease. In the myocardial microenvironment, parasite antigens and host inflammatory mediators may aggravate tissue damage. The glycoinositolphospholipid (GIPL) from T. cruzi is an inflammation-eliciting antigen recognized by Toll-like receptor 4 (TLR4), whereas the proinflammatory cytokine macrophage migration inhibitory factor (MIF) promotes progression of chronic Chagas cardiomyopathy. We herein aimed to examine the involvement of GIPL and MIF in molecular mechanisms leading to a pathogenic inflammatory response in HL-1 cardiomyocytes and HMEC microvascular endothelial cells. Immunofluorescence analysis revealed that GIPL enhanced TLR4 expression in both cell types. We found that TLR4/GIPL interaction and MIF activity modulated the arachidonic acid pathway implicated in persistent inflammation. The combination of GIPL at 50 µg/ml and MIF at 50 ng/ml upregulated type 2 cyclooxygenase (COX-2) levels in HL-1 and HMEC cells, in a stronger way than each molecule acting independently. Moreover, increased expression of prostanoid synthases and release of prostaglandin E2 (PGE2) and thromboxane B2 (TxB2) were detected in stimulated cells. Transfection experiments in HL-1 and HMEC cells showed that COX-2 induction was transcriptionally regulated through GIPL-TLR4 engagement and NFκB signaling cascade. (GIPL + MIF)-triggered NFκB activation was markedly attenuated by treatment with 100 µM Fenofibrate, a PPAR-α ligand. Fenofibrate reduced COX-2-dependent generation of bioactive lipids in HL-1 and HMEC cells. In addition, Fenofibrate abolished (GIPL + MIF)-fostered release of NO, IL-1ß, IL-6, TNF-α, and CCL2. The combined actions of GIPL and MIF display potential for amplifying the inflammatory response in myocardium of parasite-infected hosts. Our current findings might help develop more effective measures to ameliorate cardiovascular abnormalities associated with Chagas heart disease.


Subject(s)
Chagas Disease , Fenofibrate , Macrophage Migration-Inhibitory Factors , Trypanosoma cruzi , Humans , Macrophage Migration-Inhibitory Factors/metabolism , Toll-Like Receptor 4 , Myocytes, Cardiac/metabolism , Cyclooxygenase 2 , Endothelial Cells/metabolism , Inflammation
2.
Front Immunol ; 12: 782891, 2021.
Article in English | MEDLINE | ID: mdl-34925364

ABSTRACT

Benznidazole (Bzl), the drug of choice in many countries for the treatment of Chagas disease, leads to parasite clearance in the early stages of infection and contributes to immunomodulation. In addition to its parasiticidal effect, Bzl inhibits the NF-κB pathway. In this regard, we have previously described that this occurs through IL-10/STAT3/SOCS3 pathway. PI3K pathway is involved in the regulation of the immune system by inhibiting NF-κB pathway through STAT3. In this work, the participation of PI3K in the immunomodulatory effects of Bzl in cardiac and immune cells, the main targets of Chagas disease, was further studied. For that, we use a murine primary cardiomyocyte culture and a monocyte/macrophage cell line (RAW 264.7), stimulated with LPS in presence of LY294002, an inhibitor of PI3K. Under these conditions, Bzl could neither increase SOCS3 expression nor inhibit the NOS2 mRNA expression and the release of NOx, both in cardiomyocytes and macrophages. Macrophages are crucial in the development of Chronic Chagas Cardiomyopathy. Thus, to deepen our understanding of how Bzl acts, the expression profile of M1-M2 macrophage markers was evaluated. Bzl inhibited the release of NOx (M1 marker) and increased the expression of Arginase I (M2 marker) and a negative correlation was found between them. Besides, LPS increased the expression of pro-inflammatory cytokines. Bzl treatment not only inhibited this effect but also increased the expression of typical M2-macrophage markers like Mannose Receptor, TGF-ß, and VEGF-A. Moreover, Bzl increased the expression of PPAR-γ and PPAR-α, known as key regulators of macrophage polarization. PI3K directly regulates M1-to-M2 macrophage polarization. Since p110δ, catalytic subunit of PI3Kδ, is highly expressed in immune cells, experiments were carried out in presence of CAL-101, a specific inhibitor of this subunit. Under this condition, Bzl could neither increase SOCS3 expression nor inhibit NF-κB pathway. Moreover, Bzl not only failed to inhibit the expression of pro-inflammatory cytokines (M1 markers) but also could not increase M2 markers. Taken together these results demonstrate, for the first time, that the anti-inflammatory effect of Bzl depends on PI3K activity in a cell line of murine macrophages and in primary culture of neonatal cardiomyocytes. Furthermore, Bzl-mediated increase expression of M2-macrophage markers involves the participation of the p110δ catalytic subunit of PI3Kδ.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chagas Cardiomyopathy/drug therapy , Class I Phosphatidylinositol 3-Kinases/metabolism , Nitroimidazoles/pharmacology , Animals , Animals, Newborn , Anti-Inflammatory Agents/therapeutic use , Chagas Cardiomyopathy/immunology , Chromones/pharmacology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Disease Models, Animal , Female , Humans , Lipopolysaccharides/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Morpholines/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Nitroimidazoles/therapeutic use , Primary Cell Culture , RAW 264.7 Cells
3.
Front Cell Infect Microbiol ; 11: 785166, 2021.
Article in English | MEDLINE | ID: mdl-35360222

ABSTRACT

Chronic Chagas disease cardiomyopathy (CCC) is the most important clinical manifestation of infection with Trypanosma cruzi (T. cruzi) due to its frequency and effects on morbidity and mortality. Peripheral blood mononuclear cells (PBMC) infiltrate the tissue and differentiate into inflammatory macrophages. Advances in pathophysiology show that myeloid cell subpopulations contribute to cardiac homeostasis, emerging as possible therapeutic targets. We previously demonstrated that fenofibrate, PPARα agonist, controls inflammation, prevents fibrosis and improves cardiac function in a murine infection model. In this work we investigated the spontaneous release of inflammatory cytokines and chemokines, changes in the frequencies of monocyte subsets, and fenofibrate effects on PBMC of seropositive patients with different clinical stages of Chagas disease. The results show that PBMC from Chagas disease patients display higher levels of IL-12, TGF-ß, IL-6, MCP1, and CCR2 than cells from uninfected individuals (HI), irrespectively of the clinical stage, asymptomatic (Asy) or with Chagas heart disease (CHD). Fenofibrate reduces the levels of pro-inflammatory mediators and CCR2 in both Asy and CHD patients. We found that CHD patients display a significantly higher percentage of classical monocytes in comparison with Asy patients and HI. Besides, Asy patients have a significantly higher percentage of non-classical monocytes than CHD patients or HI. However, no difference in the intermediate monocyte subpopulation was found between groups. Moreover, monocytes from Asy or CHD patients exhibit different responses upon stimulation in vitro with T. cruzi lysates and fenofibrate treatment. Stimulation with T. cruzi significantly increases the percentage of classical monocytes in the Asy group whereas the percentage of intermediate monocytes decreases. Besides, there are no changes in their frequencies in CHD or HI. Notably, stimulation with T. cruzi did not modify the frequency of the non-classical monocytes subpopulation in any of the groups studied. Moreover, fenofibrate treatment of T. cruzi-stimulated cells, increased the frequency of the non-classical subpopulation in Asy patients. Interestingly, fenofibrate restores CCR2 levels but does not modify HLA-DR expression in any groups. In conclusion, our results emphasize a potential role for fenofibrate as a modulator of monocyte subpopulations towards an anti-inflammatory and healing profile in different stages of chronic Chagas disease.


Subject(s)
Chagas Disease , Fenofibrate , Animals , Cytokines/metabolism , Fenofibrate/metabolism , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Humans , Leukocytes, Mononuclear/metabolism , Mice , Monocytes/metabolism
4.
Front Immunol ; 11: 572178, 2020.
Article in English | MEDLINE | ID: mdl-33072115

ABSTRACT

IL-10 is an anti-inflammatory cytokine that plays a significant role in the modulation of the immune response in many pathological conditions, including infectious diseases. Infection with Trypanosoma cruzi (T. cruzi), the etiological agent of Chagas disease, results in an ongoing inflammatory response that may cause heart dysfunction, ultimately leading to heart failure. Given its infectious and inflammatory nature, in this work we analyzed whether the lack of IL-10 hinders the anti-inflammatory effects of fenofibrate, a PPARα ligand, in a murine model of Chagas heart disease (CHD) using IL-10 knockout (IL-10 KO) mice. Our results show fenofibrate was able to restore the abnormal cardiac function displayed by T. cruzi-infected mice lacking IL-10. Treatment with fenofibrate reduced creatine kinase (CK) levels in sera of IL-10 KO mice infected with T. cruzi. Moreover, although fenofibrate could not modulate the inflammatory infiltrates developing in the heart, it was able to reduce the increased collagen deposition in infected IL-10 KO mice. Regarding pro-inflammatory mediators, the most significant finding was the increase in serum IL-17. These were reduced in IL-10 KO mice upon fenofibrate treatment. In agreement with this, the expression of RORγt was reduced. Infection of IL-10 KO mice increased the expression of YmI, FIZZ and Mannose Receptor (tissue healing markers) that remained unchanged upon treatment with fenofibrate. In conclusion, our work emphasizes the role of anti-inflammatory mechanisms to ameliorate heart function in CHD and shows, for the first time, that fenofibrate attains this through IL-10-dependent and -independent mechanisms.


Subject(s)
Chagas Cardiomyopathy/drug therapy , Fenofibrate/therapeutic use , Hypolipidemic Agents/therapeutic use , Interleukin-10/metabolism , Myocardium/pathology , Trypanosoma cruzi/physiology , Trypanosomiasis/drug therapy , Animals , Cells, Cultured , Chagas Cardiomyopathy/immunology , Creatine Kinase/blood , Disease Models, Animal , Humans , Interleukin-10/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Trypanosomiasis/immunology , Wound Healing
5.
Front Immunol ; 10: 1267, 2019.
Article in English | MEDLINE | ID: mdl-31214200

ABSTRACT

Anti-parasitic treatment for Chagas disease mainly relies on benznidazole, which is virtually the only drug available in the market. Besides its anti-parasitic effects, benznidazole has anti-inflammatory properties. In this work we studied the mechanisms involved in the latter, demonstrating the participation of the IL-10/STAT3/SOCS3 pathway. To achieve this goal, the anti-inflammatory properties of benznidazole were studied using an in vitro model of cardiomyocyte primary culture stimulated with LPS. LPS increased both SOCS3 expression and STAT3 phosphorylation. The addition of benznidazole increased their expression even further. Specific inhibition of STAT3 precluded this effect, suggesting a role for STAT3 in the increase of SOCS3 expression induced by benznidazole. To assess the participation of SOCS3 in the anti-inflammatory effect of benznidazole, we accomplished specific knockdown of SOCS3 with siRNA. Silencing of SOCS3 in cardiomyocytes precluded the inhibitory effects of benznidazole on TNF-α, IL-6, iNOS expression and NO release. Moreover, in the absence of SOCS3, benznidazole could neither prevent IKK phosphorylation nor IκBα degradation, supporting the notion that SOCS3 is required for the benznidazole-mediated inhibition of the NF-κB pathway. Previously, we demonstrated that IL-10 increases the expression of SOCS3 in cultured cardiomyocytes. Here, we found that benznidazole shows a trend to increased IL-10 expression. To evaluate whether benznidazole increased SOCS3 in an IL-10-dependent manner, cardiomyocytes from IL-10 knockout mice were pre-treated with benznidazole and stimulated with LPS. Benznidazole neither inhibited NO release nor avoid IKK phosphorylation or IκBα degradation, showing that IL-10 is required for benznidazole-mediated inhibition of NF-κB. Moreover, exogenous addition of IL-10 to IL-10 knockout cardiomyocytes restored the inhibitory effect of benznidazole on NO release. The results reported herein show, for the first time, that the IL-10/STAT3/SOCS3 axis is involved in the anti-inflammatory effects of benznidazole. These findings may add up to new therapeutic strategies for chronic Chagas disease given its inflammatory nature.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Interleukin-10/metabolism , Nitroimidazoles/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Biomarkers , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation , Inflammation Mediators/metabolism , Interleukin-10/genetics , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitroimidazoles/chemistry , Suppressor of Cytokine Signaling 3 Protein/genetics
6.
Int J Parasitol Drugs Drug Resist ; 7(3): 378-387, 2017 12.
Article in English | MEDLINE | ID: mdl-29040909

ABSTRACT

Trypanosoma cruzi induces serious cardiac alterations during the chronic infection. Intense inflammatory response observed from the beginning of infection, is critical for the control of parasite proliferation and evolution of Chagas disease. Peroxisome proliferator-activated receptors (PPAR)-α, are known to modulate inflammation. In this study we investigated whether a PPAR-α agonist, Fenofibrate, improves cardiac function and inflammatory parameters in a murine model of T. cruzi infection. BALB/c mice were sequentially infected with two T. cruzi strains of different genetic background. Benznidazole, commonly used as trypanocidal drug, cleared parasites but did not preclude cardiac pathology, resembling what is found in human chronic chagasic cardiomyopathy. Fenofibrate treatment restored to normal values the ejection and shortening fractions, left ventricular end-diastolic, left ventricular end-systolic diameter, and isovolumic relaxation time. Moreover, it reduced cardiac inflammation and fibrosis, decreased the expression of pro-inflammatory (IL-6, TNF-α and NOS2) and heart remodeling mediators (MMP-9 and CTGF), and reduced serum creatine kinase activity. The fact that Fenofibrate partially inhibited NOS2 expression and NO release in the presence of a PPAR-α non-competitive inhibitor, suggested it also acted through PPAR-α-independent pathways. Since IκBα cytosolic degradation was inhibited by Fenofibrate, it can be concluded that the NFκB pathway has a role in its effects. Thus, we demonstrate that Fenofibrate acts through PPAR-α-dependent and -independent pathways. Our study shows that combined treatment with Fenofibrate plus Benznidazole is able both to reverse the cardiac dysfunction associated with the ongoing inflammatory response and fibrosis and to attain parasite clearance in an experimental model of Chagas disease.


Subject(s)
Chagas Cardiomyopathy/drug therapy , Fenofibrate/therapeutic use , Nitroimidazoles/therapeutic use , Trypanocidal Agents/therapeutic use , Ventricular Dysfunction/drug therapy , Animals , Chagas Cardiomyopathy/complications , Chagas Cardiomyopathy/parasitology , Chagas Disease/complications , Chagas Disease/drug therapy , Chagas Disease/parasitology , Diastole/drug effects , Fenofibrate/administration & dosage , Fibrosis/drug therapy , Humans , Inflammation/drug therapy , Inflammation/parasitology , Inflammation/physiopathology , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/drug effects , Nitric Oxide Synthase Type II/drug effects , Nitroimidazoles/administration & dosage , Nitroimidazoles/adverse effects , PPAR alpha/agonists , Stroke Volume/drug effects , Trypanocidal Agents/administration & dosage , Trypanocidal Agents/adverse effects , Trypanosoma cruzi/drug effects , Tumor Necrosis Factor-alpha/drug effects , Ventricular Dysfunction/etiology , Ventricular Function/drug effects
7.
Biochim Biophys Acta ; 1852(5): 893-904, 2015 May.
Article in English | MEDLINE | ID: mdl-25557389

ABSTRACT

Trypanosoma cruzi, the etiological agent of Chagas' disease, induces a persistent inflammatory response. Macrophages are a first line cell phenotype involved in the clearance of infection. Upon parasite uptake, these cells increase inflammatory mediators like NO, TNF-α, IL-1ß and IL-6, leading to parasite killing. Although desired, inflammatory response perpetuation and exacerbation may lead to tissue damage. Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent nuclear transcription factors that, besides regulating lipid and carbohydrate metabolism, have a significant anti-inflammatory effect. This is mediated through the interaction of the receptors with their ligands. PPARγ, one of the PPAR isoforms, has been implicated in macrophage polarization from M1, the classically activated phenotype, to M2, the alternatively activated phenotype, in different models of metabolic disorders and infection. In this study, we show for the first time that, besides PPARγ, PPARα is also involved in the in vitro polarization of macrophages isolated from T. cruzi-infected mice. Polarization was evidenced by a decrease in the expression of NOS2 and proinflammatory cytokines and the increase in M2 markers like Arginase I, Ym1, mannose receptor and TGF-ß. Besides, macrophage phagocytic activity was significantly enhanced, leading to increased parasite load. We suggest that modulation of the inflammatory response by both PPARs might be due, at least in part, to a change in the profile of inflammatory macrophages. The potential use of PPAR agonists as modulators of overt inflammatory response during the course of Chagas' disease deserves further investigation.


Subject(s)
Chagas Disease/metabolism , Macrophages/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , Animals , Arginase/genetics , Arginase/metabolism , Blotting, Western , Cells, Cultured , Chagas Disease/genetics , Chagas Disease/parasitology , Cytokines/genetics , Cytokines/metabolism , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Lectins/genetics , Lectins/metabolism , Ligands , Macrophage Activation/drug effects , Macrophages/classification , Macrophages/drug effects , Male , Mice, Inbred BALB C , Microscopy, Fluorescence , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , PPAR alpha/genetics , PPAR gamma/genetics , Phagocytosis/drug effects , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Pyrimidines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Trypanosoma cruzi/physiology , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
8.
PLoS One ; 8(11): e79445, 2013.
Article in English | MEDLINE | ID: mdl-24260222

ABSTRACT

Trypanosoma cruzi (T. cruzi) infection produces an intense inflammatory response which is critical for the control of the evolution of Chagas' disease. Interleukin (IL)-10 is one of the most important anti-inflammatory cytokines identified as modulator of the inflammatory reaction. This work shows that exogenous addition of IL-10 inhibited ERK1/2 and NF-κB activation and reduced inducible nitric oxide synthase (NOS2), metalloprotease (MMP) -9 and MMP-2 expression and activities, as well as tumour necrosis factor (TNF)-α and interleukin (IL)-6 expression, in T. cruzi-infected cardiomyocytes. We found that T. cruzi and IL-10 promote STAT3 phosphorylation and up-regulate the expression of suppressor of cytokine signalling (SOCS)-3 thereby preventing NF-κB nuclear translocation and ERK1/2 phosphorylation. Specific knockdown of SOCS-3 by small interfering RNA (siRNA) impeded the IL-10-mediated inhibition of NF-κB and ERK1/2 activation. As a result, the levels of studied pro-inflammatory mediators were restored in infected cardiomyocytes. Our study reports the first evidence that T. cruzi up- regulates SOCS-3 expression and highlights the relevance of IL-10 in the modulation of pro-inflammatory response of cardiomyocytes in Chagas' disease.


Subject(s)
Interleukin-10/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/parasitology , NF-kappa B/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Trypanosoma cruzi/pathogenicity , Animals , Cells, Cultured , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Myocytes, Cardiac/drug effects , Phosphorylation/drug effects
9.
J Cell Physiol ; 228(7): 1584-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23335284

ABSTRACT

Macrophages (Mps) can exert the defense against invading pathogens. During sepsis, bacterial lipopolisaccharide (LPS) activates the production of inflammatory mediators by Mps. Nitric oxide synthase (NOS) derived-nitric oxide (NO) is one of them. Besides, Mps may produce pro-angiogenic molecules such as vascular endothelial growth factor-A (VEGF-A) and metalloproteinases (MMPs). The mechanisms involved in the cardiac neovascular response by Mps during sepsis are not completely known. We investigated the ability of LPS-treated Mps from septic mice to modulate the behavior of cardiac cells as producers of NO and angiogenic molecules. In vivo LPS treatment (0.1 mg/mouse) increased NO production more than fourfold and induced de novo NOS2 expression in Mps. Immunoblotting assays also showed an induction in VEGF-A and MMP-9 expression in lysates obtained from LPS-treated Mps, and MMP-9 activity was detected by zymography in cell supernatants. LPS-activated Mps co-cultured with normal heart induced the expression of CD31 and VEGF-A in heart homogenates and increased MMP-9 activity in the supernatants. By immunohistochemistry, we detected new blood vessel formation in hearts cultured with LPS treated Mps. When LPS-stimulated Mps were co-cultured with isolated cardiomyocytes in a transwell assay, the expression of NOS2, VEGF-A and MMP-9 was induced in cardiac cells. In addition, MMP-9 activity was up-regulated in the supernatant of cardiomyocytes. The latter was due to NOS2 induction in Mps from in vivo LPS-treated mice. In conclusion LPS-treated Mps are inducers of inflammatory/angiogenic mediators in cardiac cells, which could be triggering neovascularization, as an attempt to improve cardiac performance in sepsis.


Subject(s)
Macrophages, Peritoneal/metabolism , Myocardium/metabolism , Neovascularization, Pathologic/metabolism , Nitric Oxide Synthase/metabolism , Sepsis/metabolism , Animals , Female , Immunity, Innate , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Myocardium/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/immunology , Sepsis/complications , Sepsis/immunology , Vascular Endothelial Growth Factor A/metabolism
10.
Biochim Biophys Acta ; 1832(1): 239-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22917565

ABSTRACT

Trypanosoma cruzi (T. cruzi), the etiological agent of Chagas' disease, causes cardiac alterations in the host. Although the main clinical manifestations arise during the chronic stage, the mechanisms leading to heart damage develop early during infection. In fact, an intense inflammatory response is observed from acute stage of infection. Recently, peroxisome proliferator-activated receptors (PPARs) have attracted research interest due to their participation in the modulation of inflammation. In this work we addressed the role of 15-Deoxy-∆(12,14) ProstaglandinJ2 (15dPGJ2), a PPARγ natural ligand in the regulation of inflammatory mediators, in acute and chronic experimental mouse models of Chagas' disease with the RA and K98 T. cruzi strains, respectively. This work demonstrates that 15dPGJ2 treatment inhibits the expression and activity of inducible nitric oxide synthase (NOS2) as well as TNF-α and IL-6 mRNA levels. Also, expression and activity of metalloproteinases 2 (MMP-2) and 9 (MMP9) were inhibited by 15dPGJ2. Moreover GW9662, a specific PPARγ antagonist, revealed the participation of other signaling pathways since, in GW9662 presence, 15dPJG2 had a partial effect on the inhibition of inflammatory parameters in the acute model of infection. Accordingly, NF-κB activation was demonstrated, assessing p65 nuclear translocation in the hearts of infected mice with both T. cruzi strains. Such effect was inhibited after 15dPGJ2 treatment. Our findings support the concept that in vivo PPARγ and NF-κB pathways are implicated in the inhibitory effects of 15dPGJ2 on inflammatory mediators at different times depending on whether the infection is caused by the lethal or non-lethal T. cruzi strain.


Subject(s)
Chagas Disease/drug therapy , Chagas Disease/immunology , Inflammation Mediators/immunology , Myocardium/immunology , Prostaglandin D2/administration & dosage , Trypanosoma cruzi/physiology , Animals , Chagas Disease/genetics , Chagas Disease/parasitology , Down-Regulation/drug effects , Heart/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Ligands , Male , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , PPAR gamma/genetics , PPAR gamma/immunology , Trypanosoma cruzi/pathogenicity , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
PPAR Res ; 2012: 528435, 2012.
Article in English | MEDLINE | ID: mdl-22448167

ABSTRACT

Chagas disease, which is caused by Trypanosoma cruzi (T. cruzi), remains a substantial public health concern and an important cause of morbidity and mortality in Latin America. T. cruzi infection causes an intense inflammatory response in diverse tissues by triggering local expression of inflammatory mediators, which results in the upregulation of the levels of cytokines and chemokines, and important cardiac alterations in the host, being one of the most characteristic damages of Chagas disease. Therefore, controlling the inflammatory reaction becomes critical for the control of the proliferation of the parasite and of the evolution of Chagas disease. The nuclear receptors known as peroxisome proliferator-activated receptors (PPARs) have emerged as key regulators of lipid metabolism and inflammation. The precise role of PPAR ligands in T. cruzi infection or in Chagas disease is poorly understood. This review summarizes our knowledge about T. cruzi infection as well as about the activation of PPARs and the potential role of their ligands in the resolution of inflammation, with the aim to address a new pharmacological approach to improve the host health.

12.
Int J Parasitol ; 41(5): 553-62, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21215746

ABSTRACT

Trypanosoma cruzi infection produces an intense inflammatory response in diverse tissues including the heart. The inflammatory reaction is critical for the control of the parasites' proliferation and evolution of Chagas disease. 15-Deoxy-Δ(12,14) prostaglandin J(2) (15dPGJ2) can repress the inflammatory response in many experimental models. However, the precise role of peroxisome proliferator-activated receptor γ (PPARγ) ligands in T. cruzi infection or in Chagas disease is poorly understood. This work reports the first evidence that 15dPGJ2 treatment increases the number of intracellular parasites as shown by fluorescence microscopy and it is also able to inhibit the expression and activity of different inflammatory enzymes such as inducible nitric oxide synthase (NOS-2), matrix metalloproteinases 2 and 9 (MMP-2, MMP-9), as well as pro-inflammatory cytokine (TNF-α and IL-6) mRNA expression in neonatal mouse cardiomyocytes after T. cruzi infection. Transfection of cardiomyocytes with small interfering RNA (siRNA) induces silencing of PPARγ and impairs the effects of 15dPGJ2 on the modulation of pro-inflammatory enzymes. Moreover, transfection restores the ability of these cells to control the intracellular growth of T. cruzi. We also found that PPARγ-independent pathways are involved, since 15dPGJ2 also exerts its effect through extracellular signal-regulated kinases-mitogen-activated protein kinase (Erk-MAPK) and nuclear factor-κB (NF-κB). The use of specific pharmacological inhibitors confirmed these findings. Our data point out that 15dPGJ2 is a potent modulator of the inflammatory process and regulator of parasites growth through PPARγ-dependent and independent (Erk-MAPK- and NF-κB) pathways in T. cruzi infected neonatal cardiac cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Chagas Disease/drug therapy , Chagas Disease/immunology , Myocytes, Cardiac/immunology , Prostaglandin D2/analogs & derivatives , Trypanosoma cruzi/physiology , Animals , Cells, Cultured , Chagas Disease/genetics , Chagas Disease/parasitology , Cytokines/genetics , Cytokines/immunology , Down-Regulation/drug effects , Gene Expression/drug effects , Humans , Male , Mice , Myocytes, Cardiac/parasitology , PPAR gamma/genetics , PPAR gamma/immunology , Prostaglandin D2/therapeutic use , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/immunology
13.
Shock ; 34(1): 60-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19997048

ABSTRACT

Sepsis or endotoxemia produced by LPS followed by hypotension and multiorganic failure may lead to cardiac dysfunction contributing to mortality. Cardiac failure is usually associated to activation of nuclear factor kappaB (NF-kappaB) and mitogen-activated protein kinase (MAPK), which play an important role in proinflammatory enzymes expression. It has been shown that 15-deoxy-Delta12,14 prostaglandin J2 (15dPGJ2) can repress the inflammatory response by means of peroxisome proliferator-activated receptor gamma (PPARgamma)-dependent and -independent mechanisms. However, its precise role in heart is poorly understood. In the present study, mouse neonatal cardiomyocytes were isolated and stimulated with LPS to investigate the role of PPARgamma-specific ligands 15dPGJ2 and rosiglitazone on cardiac inflammatory response. Inducible NO synthase, cyclooxygenase 2, and metalloproteinase 9 mRNA levels, protein expression, and activity were inhibited with 15dPGJ2 but not by rosiglitazone. Peroxisome proliferator-activated receptor gamma antagonist, GW9662, prevented all these 15dPGJ2 actions. To go inside the mechanisms by which 15dPGJ2 exerts inhibitory effects, cells were preincubated with specific chemical inhibitors of NF-kappaB and p38 MAPK, and we found that these signaling cascades are implicated in 15dPGJ2 action as well as PPARgamma. These results suggest that only the natural PPARgamma ligand, 15dPGJ2, but not the synthetic one, rosiglitazone, regulates the inflammatory response by inhibition of inducible NO synthase, cyclooxygenase 2, and metalloproteinase 9 expression. Moreover, our results offer an additional 15dPGJ2 mechanism of action, despite PPARgamma, showing NF-kappaB and p38 MAPK participation.


Subject(s)
Cyclooxygenase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Nitric Oxide Synthase Type II/metabolism , PPAR gamma/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Thiazolidinediones/pharmacology , Animals , Animals, Newborn , Blotting, Western , Cells, Cultured , Dinoprostone/metabolism , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone
14.
Microbes Infect ; 10(14-15): 1431-9, 2008.
Article in English | MEDLINE | ID: mdl-18801455

ABSTRACT

Although dam mutants of Salmonella have been proposed as live vaccines, their capacity to trigger cell inflammatory cascades has not been fully elucidated. We investigated in detail the ability of Salmonella enterica dam mutant to activate the signalling pathways of the inflammatory response in RAW 264.7 cells. Apoptosis in macrophages treated with Salmonella dam mutant was low. Similarly, the expression of both NOS-2 and COX-2 and subsequently the production of NO and PGE(2) was significantly reduced. Also, Salmonella dam mutant induced an attenuated activation of the inflammatory signalling pathway as indicated by the reduced degradation of IkappaBalpha and IkappaBbeta and the low IkappaBalpha phosphorylation found. In addition, translocation of p65 to the nucleus was notably impaired and the amount of phosphorylated p44, p42 and p38 MAPKs was clearly reduced in extracts from dam-infected macrophages. These results indicate that the lack of ERK and p38 phosphorylation at the proper time in dam-infected cells notably reduces the engagement of subsequent signalling pathways involved in the full activation of NF-kappaB in response to infection. Taken together, these results suggest that Salmonella activation of both signalling cascades in the inflammatory response is a mechanism requiring Dam protein participation.


Subject(s)
Cyclooxygenase 2/biosynthesis , Macrophages/immunology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Salmonella enteritidis/genetics , Salmonella enteritidis/immunology , Site-Specific DNA-Methyltransferase (Adenine-Specific)/deficiency , Animals , Bacterial Proteins/genetics , Cell Line , Dinoprostone/metabolism , Mice , Nitric Oxide/metabolism
15.
FEMS Microbiol Lett ; 223(2): 231-8, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12829292

ABSTRACT

Local induction of inducible nitric oxide synthase (iNOS) and apoptosis was examined in the intestine of mice infected with virulent Salmonella enterica serovar Enteritidis 5694 (S. enteritidis) and its attenuated derivative mutant E/1/3. Both, intestinal iNOS mRNA expression and iNOS activity showed a peak at 4 h only in animals receiving the virulent S. enteritidis. Aminoguanidine treatment abrogated intestinal epithelial damage produced by virulent S. enteritidis and diminished apoptosis at the tips of the villi. Unlike the virulent strain, mutant E/1/3 induced massive iNOS expression in Peyer's patches, these findings may be related to its protective capacity. Our results suggest that intestinal iNOS participates in the early response to intestinal infection and that the final effect depends on the nature of the insult.


Subject(s)
Ileum/microbiology , Nitric Oxide Synthase/metabolism , Salmonella Infections/metabolism , Salmonella enteritidis/pathogenicity , Animals , Ileum/enzymology , Ileum/pathology , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Peyer's Patches/immunology , RNA, Messenger/analysis , Salmonella Infections/immunology , Salmonella Infections/pathology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...