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










Publication year range
3.
J Immunol ; 205(10): 2795-2805, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33037139

ABSTRACT

Hemolysis causes an increase of intravascular heme, oxidative damage, and inflammation in which macrophages play a critical role. In these cells, heme can act as a prototypical damage-associated molecular pattern, inducing TLR4-dependent cytokine production through the MyD88 pathway, independently of TRIF. Heme promotes reactive oxygen species (ROS) generation independently of TLR4. ROS and TNF production contribute to heme-induced necroptosis and inflammasome activation; however, the role of ROS in proinflammatory signaling and cytokine production remains unknown. In this study, we demonstrate that heme activates at least three signaling pathways that contribute to a robust MAPK phosphorylation and cytokine expression in mouse macrophages. Although heme did not induce a detectable Myddosome formation, the TLR4/MyD88 axis was important for phosphorylation of p38 and secretion of cytokines. ROS generation and spleen tyrosine kinase (Syk) activation induced by heme were critical for most proinflammatory signaling pathways, as the antioxidant N-acetyl-l-cysteine and a Syk inhibitor differentially blocked heme-induced ROS, MAPK phosphorylation, and cytokine production in macrophages. Early generated mitochondrial ROS induced by heme was Syk dependent, selectively promoted the phosphorylation of ERK1/2 without affecting JNK or p38, and contributed to CXCL1 and TNF production. Finally, lethality caused by sterile hemolysis in mice required TLR4, TNFR1, and mitochondrial ROS, supporting the rationale to target these pathways to mitigate tissue damage of hemolytic disorders.


Subject(s)
Heme/metabolism , Hemolysis/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , Animals , Chemokine CXCL1/metabolism , Disease Models, Animal , Humans , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/immunology , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Syk Kinase/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
PLoS Pathog ; 16(7): e1008599, 2020 07.
Article in English | MEDLINE | ID: mdl-32692767

ABSTRACT

Heme oxygenase (HO-1) mediates the enzymatic cleavage of heme, a molecule with proinflammatory and prooxidant properties. HO-1 activity deeply impacts host capacity to tolerate infection through reduction of tissue damage or affecting resistance, the ability of the host to control pathogen loads. In this Review, we will discuss the contribution of HO-1 in different and complex protozoan infections, such as malaria, leishmaniasis, Chagas disease, and toxoplasmosis. The complexity of these infections and the pleiotropic effects of HO-1 constitute an interesting area of study and an opportunity for drug development.


Subject(s)
Heme Oxygenase-1/metabolism , Protozoan Infections/enzymology , Animals , Humans , Immune Tolerance/physiology
5.
Clin Rev Allergy Immunol ; 58(1): 15-24, 2020 Feb.
Article in English | MEDLINE | ID: mdl-30680604

ABSTRACT

Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine that participates in innate and adaptive immune responses. MIF contributes to the resistance against infection agents, but also to the cellular and tissue damage in infectious, autoimmune, and allergic diseases. In the past years, several studies demonstrated a critical role for MIF in the pathogenesis of type-2-mediated inflammation, including allergy and helminth infection. Atopic patients have increased MIF amounts in affected tissues, mainly produced by immune cells such as macrophages, Th2 cells, and eosinophils. Increased MIF mRNA and protein are found in activated Th2 cells, while eosinophils stock pre-formed MIF protein and secrete high amounts of MIF upon stimulation. In mouse models of allergic asthma, the lack of MIF causes an almost complete abrogation of the cardinal signs of the disease including mucus secretion, eosinophilic inflammation, and airway hyper-responsiveness. Additionally, blocking the expression of MIF in animal models leads to significant reduction of pathological signs of eosinophilic inflammation such as rhinitis, atopic dermatitis, eosinophilic esophagitis and helminth infection. A number of studies indicate that MIF is important in the effector phase of type-2 immune responses, while its contribution to Th2 differentiation and IgE production is not consensual. MIF has been found to intervene in different aspects of eosinophil physiology including differentiation, survival, activation, and migration. CD4+ T cells and eosinophils express CD74 and CXCR4, receptors able to signal upon MIF binding. Blockage of these receptors with neutralizing antibodies or small molecule antagonists also succeeds in reducing the signals of inflammation in experimental allergic models. Together, these studies demonstrate an important contribution of MIF on eosinophil biology and in the pathogenesis of allergic diseases and helminth infection.


Subject(s)
Disease Susceptibility , Eosinophils/immunology , Eosinophils/metabolism , Inflammation/etiology , Inflammation/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Animals , Biomarkers , Bone Marrow/metabolism , Bone Marrow/pathology , Eosinophils/pathology , Host-Parasite Interactions , Host-Pathogen Interactions , Humans , Hypersensitivity/etiology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Inflammation/pathology , Signal Transduction
6.
Article in English | MEDLINE | ID: mdl-31297344

ABSTRACT

Leishmaniasis is an infectious disease caused by protozoans of the genus Leishmania. The macrophage is the resident cell in which the parasite replicates and it is important to identify new compounds that can aid in parasite elimination since the drugs used to treat leishmaniasis are toxic and present side effects. We have previously shown that treatment of Leishmania braziliensis-infected macrophages with DETC (Diethyldithiocarbamate) induces parasite killing, in vivo. Thus, the objective of this study was to further evaluate the effect of oxidants and antioxidants in L. braziliensis-infected macrophages, following treatment with either oxidizing Hydrogen Peroxide, Menadione, DETC, or antioxidant [NAC (N-Acetyl-Cyteine), Apocynin, and Tempol] compounds. We determined the percentage of infected macrophages and number of amastigotes. Promastigote survival was also evaluated. Both DETC (SOD-inhibitor) and Tempol (SOD-mimetic) decreased the percentage of infected cells and parasite load. Hydrogen peroxide did not interfere with parasite burden, while superoxide-generator Menadione had a reducing effect. On the other hand, NAC (GSH-replenisher) and Apocynin (NADPH-oxidase inhibitor) increased parasite burden. Tempol surfaces as an interesting candidate for the chemotherapy of CL with an IC50 of 0.66 ± 0.08 mM and selectivity index of 151. While it remains obscure how a SOD-mimetic may induce leishmanicidal effects, we suggest the possibility of developing Tempol-based topical applications for the treatment of cutaneous leishmaniasis caused by L. braziliensis.


Subject(s)
Cyclic N-Oxides/pharmacology , Leishmania braziliensis/drug effects , Leishmaniasis, Cutaneous/drug therapy , Macrophages/drug effects , Superoxide Dismutase/pharmacology , Acetophenones/pharmacology , Animals , Antioxidants , Disease Models, Animal , Ditiocarb , Drug Therapy/methods , Female , Hydrogen Peroxide , Inhibitory Concentration 50 , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Parasite Load , Spin Labels , Vitamin K 3/pharmacology
7.
J Mol Cell Cardiol ; 131: 101-111, 2019 06.
Article in English | MEDLINE | ID: mdl-31029578

ABSTRACT

AIMS: Cardiac arrhythmias are one of the most important remote complications after kidney injury. Renal ischemia reperfusion (I/R) is a major cause of acute renal injury predisposing to several remote dysfunctions, including cardiac electrical disturbance. Since IL-1ß production dependent on NLRP3 represents a link between tissue malfunctioning and cardiac arrhythmias, here we tested the hypothesis that longer ventricular repolarization and arrhythmias after renal I/R depend on this innate immunity sensor. METHODS AND RESULTS: Nlrp3-/- and Casp1-/- mice reacted to renal I/R with no increase in plasma IL-1ß, different from WT (wild-type) I/R. A prolonged QJ interval and an increased susceptibility to ventricular arrhythmias were found after I/R compared to Sham controls in wild-type mice at 15 days post-perfusion, but not in Nlrp3-/- or CASP1-/- I/R, indicating that the absence of NLRP3 or CASP1 totally prevented longer QJ interval after renal I/R. In contrast with WT mice, we found no renal atrophy and no renal dysfunction in Nlrp3-/- and Casp1-/- mice after renal I/R. Depletion of macrophages in vivo after I/R and a day before IL-1ß peak (at 7 days post-perfusion) totally prevented prolongation of QJ interval, suggesting that macrophages might participate as sensors of tissue injury. Moreover, treatment of I/R-WT mice with IL-1r antagonist (IL-1ra) from 8 to 15 days post perfusion did not interfere with renal function, but reversed QJ prolongation, prevented the increase in susceptibility to ventricular arrhythmias and rescued a close to normal duration and amplitude of calcium transient. CONCLUSION: Taken together, these results corroborate the hypothesis that IL-1ß is produced after sensing renal injury through NRLP3-CASP1, and IL-1ß on its turn triggers longer ventricular repolarization and increase susceptibility to cardiac arrhythmias. Still, they offer a therapeutic approach to treat cardiac arrhythmias that arise after renal I/R.


Subject(s)
Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Interleukin-1beta/metabolism , Kidney Diseases/complications , Kidney Diseases/metabolism , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Animals , Caspase 1/genetics , Caspase 1/metabolism , Immunity, Innate/physiology , Kidney Diseases/immunology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/immunology , Signal Transduction/physiology
8.
Shock ; 50(4): 483-492, 2018 10.
Article in English | MEDLINE | ID: mdl-30216298

ABSTRACT

Hemodynamic collapse and myocardial dysfunction are among the major causes of death in severe sepsis. The purpose of this study was to assess the role played by toll-like receptor 4 and by the NLRP3 inflammasome in the cardiac dysfunction that occurs after high-grade polymicrobial sepsis. We performed the colon ascendens stent peritonitis (CASP) surgery in Tlr4, Nlrp3, and caspase-1 mice. We also assessed for the first time the electrical heart function in the colon ascendens stent peritonitis (CASP) model. The QJ interval was increased in wild-type C57BL/6J mice after CASP when compared with sham controls, a result paralleled by an increase in the cardiac action potential (AP) duration (APD). The decreases in ejection fraction (EF), left ventricle end diastolic volume, stroke volume, and cardiac output found after CASP were similar among all groups of mice. Similar heart response was found when Nlrp3 mice were submitted to high-grade cecal ligation and puncture. Despite developing cardiac dysfunction similar to wild types after CASP, Nlrp3 mice had reduced circulating levels of interleukin (IL)-1ß, IL-6 and tumor necrosis factor-α. Our results demonstrate that the genetic ablation of Tlr4, Nlrp3, and caspase-1 does not prevent the cardiac dysfunction, despite preventing the increase in pro-inflammatory cytokines, indicating that these are not feasible targets to therapy in high-grade sepsis.


Subject(s)
Caspase 1/metabolism , Colon/metabolism , Heart Diseases/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peritonitis/complications , Peritonitis/metabolism , Toll-Like Receptor 4/metabolism , Animals , Echocardiography , Male , Mice , Mice, Inbred C57BL , Ventricular Function, Left/physiology
9.
PLoS Pathog ; 14(4): e1006928, 2018 04.
Article in English | MEDLINE | ID: mdl-29672619

ABSTRACT

The activation of macrophage respiratory burst in response to infection with Trypanosoma cruzi inflicts oxidative damage to the host's tissues. For decades, the role of reactive oxygen species (ROS) in the elimination of T. cruzi was taken for granted, but recent evidence suggests parasite growth is stimulated in oxidative environments. It is still a matter of debate whether indeed oxidative environments provide ideal conditions (e.g., iron availability in macrophages) for T. cruzi growth and whether indeed ROS signals directly to stimulate growth. Nitric oxide (NO) and ROS combine to form peroxynitrite, participating in the killing of phagocytosed parasites by activated macrophages. In response to infection, mitochondrial ROS are produced by cardiomyocytes. They contribute to oxidative damage that persists at the chronic stage of infection and is involved in functional impairment of the heart. In this review, we discuss how oxidative stress helps parasite growth during the acute stage and how it participates in the development of cardiomyopathy at the chronic stage.


Subject(s)
Chagas Disease/complications , Heart Diseases/etiology , Macrophages/microbiology , Oxidative Stress , Reactive Oxygen Species/metabolism , Trypanosoma cruzi/pathogenicity , Animals , Chagas Disease/microbiology , Humans
10.
Int J Cardiol ; 240: 354-359, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28320606

ABSTRACT

BACKGROUND: Cardiac arrhythmias are one of the main causes of death in ChCP and other dilated cardiomyopathies. Previous studies demonstrated that ventricular arrhythmias are associated with the presence of autoantibodies with beta-adrenergic activity, Ab-ß. OBJECTIVES: The aim of this study was to investigate whether Ab-ß, present in chronic chagasic patients (ChCP), induce cardiac arrhythmias in the pharmacological type-2 long QT syndrome model (LQTS-2). METHODS/RESULTS: The LQTS2 was established by perfusion of Tyrode saline solution with a potassium channel blocker E-4031 (5µM) in isolated rabbit hearts or in rabbit cardiac strips, in order to record ECG or action potential, respectively. Autoantibodies from ChCP activating (Ab-ß) or not (Ab-NR) cardiac beta 1-adrenergic receptors were used. Ab-ß, but not Ab-NR, were able to significantly shorten QT, QTc and increase Tpeak-Tend interval in the LQTS-2. A positive correlation between higher QTc and Tpeak-Tend was found after Ab-ß perfusion in the same model. In addition, in the LQTS-2 model, in almost 75% (11/15) of the hearts perfused with Ab-ß, ventricular and atrio-ventricular electrical disturbances were observed. Atenolol abolished all Ab-ß-induced arrhythmias. Ab-ß, when perfused in a cellular LQTS-2, drastically reduced the action potential duration and evoked early afterdepolarization (EAD's), while Ab-NR did not modulate the AP properties in the LQTS-2. CONCLUSION: The results indicate that Ab-ß were able to induce cardiac arrhythmias and EAD's. This phenomenon can explain, at least in part, the cellular mechanism of Ab-ß-induced arrhythmias. Furthermore, atenolol is effective for the treatment of Ab-ß-induced arrhythmias.


Subject(s)
Arrhythmias, Cardiac/blood , Arrhythmias, Cardiac/physiopathology , Autoantibodies/blood , Chagas Cardiomyopathy/blood , Long QT Syndrome/physiopathology , Receptors, Adrenergic, beta-1/blood , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anti-Arrhythmia Agents/pharmacology , Anti-Arrhythmia Agents/therapeutic use , Anti-Arrhythmia Agents/toxicity , Arrhythmias, Cardiac/etiology , Electrocardiography/drug effects , Electrocardiography/methods , Heart/drug effects , Heart/physiopathology , Humans , Long QT Syndrome/chemically induced , Longitudinal Studies , Male , Rabbits , Retrospective Studies
11.
Nat Commun ; 7: 13344, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27882934

ABSTRACT

Diabetes mellitus (DM) encompasses a multitude of secondary disorders, including heart disease. One of the most frequent and potentially life threatening disorders of DM-induced heart disease is ventricular tachycardia (VT). Here we show that toll-like receptor 2 (TLR2) and NLRP3 inflammasome activation in cardiac macrophages mediate the production of IL-1ß in DM mice. IL-1ß causes prolongation of the action potential duration, induces a decrease in potassium current and an increase in calcium sparks in cardiomyocytes, which are changes that underlie arrhythmia propensity. IL-1ß-induced spontaneous contractile events are associated with CaMKII oxidation and phosphorylation. We further show that DM-induced arrhythmias can be successfully treated by inhibiting the IL-1ß axis with either IL-1 receptor antagonist or by inhibiting the NLRP3 inflammasome. Our results establish IL-1ß as an inflammatory connection between metabolic dysfunction and arrhythmias in DM.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Interleukin-1beta/immunology , Macrophages/immunology , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Tachycardia, Ventricular/immunology , Toll-Like Receptor 2/immunology , Action Potentials , Animals , Antirheumatic Agents/pharmacology , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Caspase 1/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Inflammasomes/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Transgenic , Myocardial Contraction , Myocytes, Cardiac/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Potassium/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/metabolism , Toll-Like Receptor 2/genetics
12.
PLoS Pathog ; 12(10): e1005947, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27788262

ABSTRACT

Chronic chagasic cardiomyopathy (CCC) develops years after acute infection by Trypanosoma cruzi and does not improve after trypanocidal therapy, despite reduction of parasite burden. During disease, the heart undergoes oxidative stress, a potential causative factor for arrhythmias and contractile dysfunction. Here we tested whether antioxidants/ cardioprotective drugs could improve cardiac function in established Chagas heart disease. We chose a model that resembles B1-B2 stage of human CCC, treated mice with resveratrol and performed electrocardiography and echocardiography studies. Resveratrol reduced the prolonged PR and QTc intervals, increased heart rates and reversed sinus arrhythmia, atrial and atrioventricular conduction disorders; restored a normal left ventricular ejection fraction, improved stroke volume and cardiac output. Resveratrol activated the AMPK-pathway and reduced both ROS production and heart parasite burden, without interfering with vascularization or myocarditis intensity. Resveratrol was even capable of improving heart function of infected mice when treatment was started late after infection, while trypanocidal drug benznidazole failed. We attempted to mimic resveratrol's actions using metformin (AMPK-activator) or tempol (SOD-mimetic). Metformin and tempol mimicked the beneficial effects of resveratrol on heart function and decreased lipid peroxidation, but did not alter parasite burden. These results indicate that AMPK activation and ROS neutralization are key strategies to induce tolerance to Chagas heart disease. Despite all tissue damage observed in established Chagas heart disease, we found that a physiological dysfunction can still be reversed by treatment with resveratrol, metformin and tempol, resulting in improved heart function and representing a starting point to develop innovative therapies in CCC.


Subject(s)
Antioxidants/pharmacology , Chagas Cardiomyopathy/pathology , Stilbenes/pharmacology , Animals , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Female , Male , Metformin/pharmacology , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects , Resveratrol , Spin Labels
13.
Antimicrob Agents Chemother ; 58(4): 2076-83, 2014.
Article in English | MEDLINE | ID: mdl-24449779

ABSTRACT

In chronic schistosomiasis, hepatic fibrosis is linked to the portal hypertension that causes morbidity in Schistosoma mansoni infection. Silymarin (SIL) is a hepatoprotective and antioxidant medicament largely prescribed against liver diseases that has previously been shown to prevent fibrosis during acute murine schistosomiasis. Here we employed silymarin to try to reverse established hepatic fibrosis in chronic schistosomiasis. Silymarin or vehicle was administered to BALB/c mice every 48 h, starting on the 40th (80 days of treatment), 70th (50 days), or 110th (10 days) day postinfection (dpi). All mice were sacrificed and analyzed at 120 dpi. Treatment with silymarin reduced liver weight and granuloma sizes, reduced the increase in alanine aminotransferase and aspartate aminotransferase levels, and reduced the established hepatic fibrosis (assessed by hydroxyproline contents and picrosirius staining). Treatment with silymarin also reduced the levels of interleukin-13 (IL-13) in serum and increased the gamma interferon (IFN-γ)/IL-13 ratio. There was a linear correlation between IL-13 levels in serum and hydroxyproline hepatic content in both infected untreated and SIL-treated mice, with decreased IL-13 levels corresponding to decreased hydroxyproline hepatic contents. Treatment with either SIL or N-acetylcysteine reduced both proliferation of fibroblast cell lines and basal/IL-13-induced production of collagen I, indicating that besides inhibiting IL-13 production during infection, SIL antioxidant properties most likely contribute to inhibition of collagen production downstream of IL-13. These results show that silymarin interferes with fibrogenic cytokines, reduces established fibrosis, and inhibits downstream effects of IL-13 on fibrogenesis, indicating the drug as a safe and cheap treatment to liver fibrotic disease in schistosomiasis.


Subject(s)
Anthelmintics/therapeutic use , Liver Cirrhosis/drug therapy , Schistosomiasis/drug therapy , Silymarin/therapeutic use , Animals , Anthelmintics/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/blood , Female , Fluorescent Antibody Technique , Liver Cirrhosis/blood , Mice , Mice, Inbred BALB C , Schistosomiasis/blood , Silymarin/pharmacology
14.
Antioxid Redox Signal ; 20(6): 1000-37, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-23992156

ABSTRACT

Reactive oxygen species (ROS) are deadly weapons used by phagocytes and other cell types, such as lung epithelial cells, against pathogens. ROS can kill pathogens directly by causing oxidative damage to biocompounds or indirectly by stimulating pathogen elimination by various nonoxidative mechanisms, including pattern recognition receptors signaling, autophagy, neutrophil extracellular trap formation, and T-lymphocyte responses. Thus, one should expect that the inhibition of ROS production promote infection. Increasing evidences support that in certain particular infections, antioxidants decrease and prooxidants increase pathogen burden. In this study, we review the classic infections that are controlled by ROS and the cases in which ROS appear as promoters of infection, challenging the paradigm. We discuss the possible mechanisms by which ROS could promote particular infections. These mechanisms are still not completely clear but include the metabolic effects of ROS on pathogen physiology, ROS-induced damage to the immune system, and ROS-induced activation of immune defense mechanisms that are subsequently hijacked by particular pathogens to act against more effective microbicidal mechanisms of the immune system. The effective use of antioxidants as therapeutic agents against certain infections is a realistic possibility that is beginning to be applied against viruses.


Subject(s)
Reactive Oxygen Species/metabolism , Animals , Autophagy/physiology , Humans , T-Lymphocytes/metabolism
15.
J Clin Invest ; 122(7): 2531-42, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22728935

ABSTRACT

Oxidative damage contributes to microbe elimination during macrophage respiratory burst. Nuclear factor, erythroid-derived 2, like 2 (NRF2) orchestrates antioxidant defenses, including the expression of heme-oxygenase-1 (HO-1). Unexpectedly, the activation of NRF2 and HO-1 reduces infection by a number of pathogens, although the mechanism responsible for this effect is largely unknown. We studied Trypanosoma cruzi infection in mice in which NRF2/HO-1 was induced with cobalt protoporphyrin (CoPP). CoPP reduced parasitemia and tissue parasitism, while an inhibitor of HO-1 activity increased T. cruzi parasitemia in blood. CoPP-induced effects did not depend on the adaptive immunity, nor were parasites directly targeted. We also found that CoPP reduced macrophage parasitism, which depended on NRF2 expression but not on classical mechanisms such as apoptosis of infected cells, induction of type I IFN, or NO. We found that exogenous expression of NRF2 or HO-1 also reduced macrophage parasitism. Several antioxidants, including NRF2 activators, reduced macrophage parasite burden, while pro-oxidants promoted it. Reducing the intracellular labile iron pool decreased parasitism, and antioxidants increased the expression of ferritin and ferroportin in infected macrophages. Ferrous sulfate reversed the CoPP-induced decrease in macrophage parasite burden and, given in vivo, reversed their protective effects. Our results indicate that oxidative stress contributes to parasite persistence in host tissues and open a new avenue for the development of anti-T. cruzi drugs.


Subject(s)
Chagas Disease/parasitology , Oxidative Stress , Parasitemia/parasitology , Trypanosoma cruzi/physiology , Animals , Antioxidants/metabolism , Antiparasitic Agents/pharmacology , Antiparasitic Agents/therapeutic use , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Chagas Disease/drug therapy , Ferritins/genetics , Ferritins/metabolism , Ferrous Compounds/pharmacology , Heart/parasitology , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Host-Parasite Interactions , Iron/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred C57BL , Muscle, Skeletal/parasitology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Parasitemia/drug therapy , Protoporphyrins/pharmacology , Protoporphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Receptors, Immunologic/metabolism , Respiratory Burst , Trypanosoma cruzi/drug effects
16.
J Parasitol Res ; 2012: 413052, 2012.
Article in English | MEDLINE | ID: mdl-22496958

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a cytokine that plays a central role in immune and inflammatory responses. In the present paper, we discussed the participation of MIF in the immune response to protozoan parasite infections. As a general trend, MIF participates in the control of parasite burden at the expense of promoting tissue damage due to increased inflammation.

17.
J Biol Chem ; 285(43): 32844-32851, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20729208

ABSTRACT

Infectious diseases that cause hemolysis are among the most threatening human diseases, because of severity and/or global distribution. In these conditions, hemeproteins and heme are released, but whether heme affects the inflammatory response to microorganism molecules remains to be characterized. Here, we show that heme increased the lethality and cytokine secretion induced by LPS in vivo and enhanced the secretion of cytokines by macrophages stimulated with various agonists of innate immune receptors. Activation of nuclear factor κB (NF-κB) and MAPKs and the generation of reactive oxygen species were essential to the increase in cytokine production induced by heme plus LPS. This synergistic effect of heme and LPS was blocked by a selective inhibitor of spleen tyrosine kinase (Syk) and was abrogated in dendritic cells deficient in Syk. Moreover, inhibition of Syk and the downstream molecules PKC and PI3K reduced the reactive oxygen species generation by heme. Our results highlight a mechanism by which heme amplifies the secretion of cytokines triggered by microbial molecule activation and indicates possible pathways for therapeutic intervention during hemolytic infectious diseases.


Subject(s)
Heme/immunology , Immunity, Innate/physiology , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Protein-Tyrosine Kinases/immunology , Reactive Oxygen Species/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Heme/agonists , Heme/metabolism , Heme/pharmacology , Humans , Immunity, Innate/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/agonists , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/genetics , Protein Kinase C/immunology , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , Syk Kinase
18.
J Leukoc Biol ; 86(5): 1239-46, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19641038

ABSTRACT

CCL2/MCP-1 has emerged recently as a critical factor in infectious and autoimmune myocarditis. In fact, this chemokine is produced in great amounts in hearts from Trypanosoma cruzi-infected mice and is known to enhance parasite uptake and destruction by macrophages. Herein, we studied the involvement of CCL2 in tissue inflammation and resistance to T. cruzi. Infected CCL2(-/-) mice developed higher parasitemias and died earlier than WT mice. Close to their death, T. cruzi-infected CCL2(-/-) presented greater amounts of TNF, IFN-gamma, and IL-10 in plasma than WTs and clinical signs of systemic inflammatory response. Amastigote nests were more frequent in hearts and livers from infected CCL2(-/-) tissues than in WTs, and reduced numbers of leukocytes infiltrated their tissues. Leukocytes formed diffuse but not focal infiltrates in hearts from infected CCL2(-/-) mice, and perivascular cuffs could still be found in their livers. Infected CCL2(-/-) mice had smaller percentages of activated CD11b (Mac-1)+CD107b (Mac-3)+ macrophages and CD8+CD69(hi) cells among heart and liver infiltrates than WTs (flow cytometry), indicating that CCL2 controls subset migration/activation. CCL2 accumulated among focal heart infiltrates, suggesting that this chemokine is involved in retention of mononuclear cells in particular spots. Peritoneal macrophages from CCL2(-/-) mice displayed decreased trypanocidal activity. Our results demonstrate that CCL2 contributes to reduce parasite growth and indicate that it does so by controlling the distribution, cellular composition, and state of activation of inflammatory infiltrates in acute T. cruzi infection.


Subject(s)
Chagas Disease/physiopathology , Chemokine CCL2/deficiency , Chemokine CCL2/physiology , Trypanosoma cruzi/physiology , Acute Disease , Animals , Chagas Disease/genetics , Chagas Disease/pathology , Chemokine CCL2/genetics , Female , Genetic Predisposition to Disease , Heart/parasitology , Immunohistochemistry , Inflammation/parasitology , Liver/parasitology , Liver/pathology , Macrophages/parasitology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Spleen/parasitology , Spleen/pathology
19.
J Leukoc Biol ; 85(5): 855-61, 2009 May.
Article in English | MEDLINE | ID: mdl-19188484

ABSTRACT

Deposition of immune complexes (IC) triggers Fc gamma R-dependent inflammation, leading to tissue damage in rheumatoid arthritis, systemic lupus erythematous, immune glomerulonephritis, and several immune vasculitides. Evidences support a role for macrophage migration inhibitory factor (MIF) in a number of inflammatory diseases, but the triggering of its secretion and its physiopathological role upon IC deposition remain elusive. Herein, we show that human macrophages secreted MIF after IC recognition, which in turn controlled the secretion of TNF. Macrophages from Mif-/- mice produced smaller amounts of TNF when stimulated with IgG-opsonized erythrocytes than wild-type (WT) cells. Using passive reverse Arthus reaction in the peritoneum and lungs as a model for IC-induced inflammation, we demonstrated that Mif-/- mice had a milder response, observed by reduced neutrophil recruitment, vascular leakage, and secretion of TNF, MIP-2, and keratinocyte-derived chemokine compared with WT controls. Adoptive transfer of alveolar macrophages from WT to Mif-/- mice rescued pulmonary neutrophil recruitment and TNF production upon passive reverse Arthus reaction. Our study indicates that Arthus inflammatory reaction is largely dependent on MIF and poses macrophages as a source of the MIF released upon IC recognition. These results give experimental support to the proposition that blockade of MIF might constitute an adjunctive, therapeutic approach to IC disease.


Subject(s)
Arthus Reaction/immunology , Inflammation/immunology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Macrophages, Alveolar/immunology , Tumor Necrosis Factors/biosynthesis , Adoptive Transfer , Animals , Antigen-Antibody Complex/immunology , Cells, Cultured , Female , Humans , Inflammation/pathology , Lung/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, IgG/immunology
20.
FASEB J ; 23(4): 1262-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19088181

ABSTRACT

Macrophage migration inhibitory factor (MIF) participates in the pathogenesis of inflammatory diseases, including asthma, in which it enhances airway hypersensitivity and tissue eosinophilia. Herein, we investigated the role of MIF in eosinophilopoiesis and tissue eosinophilia using Schistosoma mansoni infection. MIF-deficient (Mif(-/-)) mice had similar numbers of adult worms, eggs, and granulomas compared to wild-type mice, but the size of granulomas was strikingly reduced due to smaller numbers of eosinophils. MIF did not affect the acquired response to infection, as Mif(-/-) mice produced normal amounts of Th2 cytokines and IgE. Nevertheless, recombinant MIF (rMIF) behaved as a chemoattractant for eosinophils, what could partially explain the reduced eosinophilia in infected Mif(-/-) mice. Moreover, the percentage of eosinophils was reduced in bone marrows of Mif(-/-) mice chronically infected with S. mansoni compared to wild type. Mif(-/-) had impaired eosinophilopoiesis in response to interleukin (IL)-5 and addition of rMIF to bone marrow cultures from IL-5 transgenic mice enhanced the generation of eosinophils. In the absence of MIF, eosinophil precursors were unable to survive the IL-5-supplemented cell culture, and were ingested by macrophages. Treatment with pancaspase inhibitor z-VAD or rMIF promoted the survival of eosinophil progenitors. Together, these results indicate that MIF participates in IL-5-driven maturation of eosinophils and in tissue eosinophilia associated with S. mansoni infection.


Subject(s)
Eosinophilia/immunology , Eosinophils/immunology , Interleukin-5/physiology , Macrophage Migration-Inhibitory Factors/physiology , Schistosomiasis mansoni/pathology , Animals , Eosinophilia/etiology , Eosinophilia/pathology , Eosinophils/pathology , Granuloma/etiology , Granuloma/immunology , Granuloma/pathology , Inflammation/pathology , Interleukin-5/immunology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Proteins/immunology , Schistosomiasis mansoni/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...