Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Exp Parasitol ; 248: 108491, 2023 May.
Article in English | MEDLINE | ID: mdl-36841467

ABSTRACT

Sustained interaction between the cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its functional receptor, fibroblast growth factor-inducible 14 (Fn14), has been linked to cardiovascular disorders. Chagas cardiomyopathy, elicited by Trypanosoma cruzi infection, is associated with chronic inflammation, fibrosis and hypertrophy. This study aimed to explore the involvement of the TWEAK/Fn 14 axis in development of Chagas heart disease. Parasite infection in vitro triggered Fn14 overexpression in atrial HL-1 myocytes and cardiac MCF fibroblasts. Fn14 levels were also increased in heart tissue from C57BL/6 mice at 130 days post-infection, particularly in myocytes and fibroblasts. Concurrently, TWEAK expression in circulating monocytes from this group was higher than that determined in uninfected controls. TWEAK/Fn14 interaction was functional in myocytes and fibroblasts isolated from infected hearts, leading to TNF receptor-associated factor 2 (TRAF2)-mediated activation of nuclear factor kappa B (NFκB) signaling. Ex vivo stimulation of both cell types with recombinant TWEAK for 24 h boosted the NFκB-regulated production of proinflammatory/profibrotic mediators (IL-1ß, IL-6, TNF-α, IL-8, CCL2, CCL5, MMP-2, MMP-9, ICAM-1, E-selectin) involved in chronic T. cruzi cardiomyopathy. We further evaluated the therapeutic potential of the soluble decoy receptor Fn14-Fc to interfere with TWEAK/Fn14-dependent pathogenic activity. Fn14-Fc treatment of chronically infected mice was effective in neutralizing the ligand and reverting electrocardiographic abnormalities, maladaptive inflammation, adverse remodeling and hypertrophy in myocardium. Altogether, these findings suggest that sustained TWEAK/Fn14 induction by persistent T. cruzi infection is implicated in cardiopathogenesis and make TWEAK/Fn14 axis a promising target for the treatment of chronic Chagas heart disease.


Subject(s)
Chagas Disease , Heart Diseases , Mice , Animals , Myocytes, Cardiac , TWEAK Receptor/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Inflammation , Fibroblasts , Heart Diseases/metabolism , Hypertrophy/metabolism
2.
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
3.
Microbes Infect ; 23(1): 104757, 2021.
Article in English | MEDLINE | ID: mdl-32956869

ABSTRACT

Upon infection by Trypanosoma cruzi, adipocytes adopt a clearly defined inflammatory phenotype with concomitant down-regulation of adiponectin expression, which influences the pathogenesis of Chagas heart disease. Herein, we examined how T. cruzi interferes with transcriptional regulation of adiponectin production in mouse adipocytes. The invading pathogen activates the Ca2+/calcineurin/NFATc4 signaling pathway in 3T3-L1 cells. Parasite-induced early activation of NFATc4 is involved in repressing adiponectin expression through recognition of the specific response element located at (-363 to -344) of the gene promoter. Nuclear import of dephosphorylated NFATc4 and decreased adiponectin levels were further demonstrated in white adipose tissue from acutely infected mice. Our current findings point to better clarify the complex role of adipose tissue in the modulation of inflammatory mechanisms operative during T. cruzi infection.


Subject(s)
Adipocytes/metabolism , Adiponectin/genetics , Chagas Disease/genetics , NFATC Transcription Factors/metabolism , Trypanosoma cruzi/physiology , 3T3-L1 Cells , Adipocytes/parasitology , Adiponectin/metabolism , Animals , Calcineurin/genetics , Calcineurin/metabolism , Calcium/metabolism , Chagas Disease/metabolism , Chagas Disease/parasitology , Down-Regulation , Gene Expression Regulation , Host-Parasite Interactions , Humans , Male , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , Signal Transduction , Trypanosoma cruzi/genetics
4.
Parasitol Int ; 81: 102248, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33238215

ABSTRACT

Curcumin (Cur) is a natural polyphenolic flavonoid isolated from the rhizomes of Curcuma longa. Its anti-inflammatory and cardioprotective properties are increasingly considered to have beneficial effects on the progression of cardiomyopathy associated with Chagas disease, caused by Trypanosoma cruzi. However, the Cur therapeutic limitation is its bioavailability and new Cur nanomedicine formulations are developed to overcome this obstacle. In this research, we provide evidence showing that oral therapy with a suboptimal dose of the standard parasiticidal drug benznidazole (BZ) in combination with Cur-loaded nanoparticles is capable of reducing myocardial parasite load, cardiac hypertrophy, inflammation and fibrosis in mice with long-term infection by T. cruzi. Treatment with BZ plus Cur was highly effective in downregulating myocardial expression of proinflammatory cytokines/chemokines (IL-1ß, TNF-α, IL-6, CCL5), and the level/activity of matrix metalloproteinases (MMP-2, MMP-9) and inducible enzymes (cyclooxygenase, nitric oxide synthase) implicated in leukocyte recruitment and cardiac remodeling. Oral administration of a Cur-based nanoformulation displays potential as a complementary strategy to the conventional BZ chemotherapy in the treatment of chronic Chagas heart disease.


Subject(s)
Chagas Cardiomyopathy/drug therapy , Curcumin/pharmacology , Nitroimidazoles/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/physiology , Animals , Chagas Cardiomyopathy/parasitology , Chronic Disease , Female , Male , Mice , Mice, Inbred C57BL
6.
Cell Immunol ; 335: 15-21, 2019 01.
Article in English | MEDLINE | ID: mdl-30384962

ABSTRACT

Increasing attention is given to the finding that macrophages under hypoxia are capable of controlling infection by the intracellular protozoan parasite Leishmania amazonensis. The hypoxia-inducible factor (HIF)-1α has been shown to play an essential role in this enhanced innate immune response. Our study aimed to explore the HIF-1α-dependent mechanisms leading to reduced survival of the parasites residing in macrophages under low oxygen conditions. Hypoxia triggered (P < 0.01) NADPH oxidase 2 (Nox2) expression and reactive oxygen species (ROS) production in J774 macrophages upon 24-h infection with L. amazonensis. Furthermore, increased (P < 0.01) expression levels of HIF-1α and macrophage migration inhibitory factor (MIF) were detected in the infected cells grown at 3% oxygen tension. We found that either HIF-1α silencing, Nox2 inhibition or MIF antagonism caused a significant (P < 0.05) reversal of the improved leishmanicidal activity displayed by the hypoxic phagocytes. Taken together, our current results suggest that, under conditions of limited availability of oxygen, activation of the HIF-1α/MIF axis via Nox2/ROS induction promotes killing of L. amazonensis amastigotes by macrophages. Such protective mechanism might operate in L. amazonensis-infected tissues where low oxygen levels prevail.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/immunology , Animals , Cell Hypoxia , Cell Line , Hypoxia/immunology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Innate , Intramolecular Oxidoreductases/physiology , Leishmania/immunology , Leishmania/physiology , Macrophage Migration-Inhibitory Factors/physiology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/physiology , Reactive Oxygen Species/metabolism
7.
Mem Inst Oswaldo Cruz ; 113(9): e180171, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30020318

ABSTRACT

BACKGROUND: The anti-inflammatory and cardioprotective properties of curcumin (Cur), a natural polyphenolic flavonoid isolated from the rhizomes of Curcuma longa, are increasingly considered to have beneficial effects on the progression of Chagas heart disease, caused by the protozoan parasite Trypanosoma cruzi. OBJECTIVE: To evaluate the effects of oral therapy with Cur on T. cruzi-mediated cardiovasculopathy in acutely infected mice and analyse the in vitro response of parasite-infected human microvascular endothelial cells treated with this phytochemical. METHODS: Inflammation of heart vessels from Cur-treated and untreated infected mice were analysed by histology, with benznidazole (Bz) as the reference compound. Parasitaemia was monitored by the direct method. Capillary permeability was visualised by Evans-blue assay. Myocardial ET-1, IL-6, and TNF-α mRNA expressions were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Microvascular endothelial HMEC-1 cells were infected in vitro with or without addition of Cur or Bz. Induction of the Ca2+/NFAT pathway was assessed by fluorometry, immunoblotting, and reporter assay. FINDINGS: Oral Cur therapy of recently infected mice reduced inflammatory cell infiltration of myocardial arteries without lowering parasite levels. Compared to that of the phosphate-buffered saline-receiving group, hearts from Cur-treated mice showed significantly decreased vessel inflammation scores (p < 0.001), vascular permeabilities (p < 0.001), and levels of IL-6/TNF-α (p < 0.01) and ET-1 (p < 0.05) mRNA. Moreover, Cur significantly (p < 0.05 for transcript; p < 0.01 for peptide) downregulated ET-1 secretion from infected HMEC-1 cells. Remarkably, Cur addition significantly (p < 0.05 at 27.0 µM) interfered with T. cruzi-dependent activation of the Ca2+/NFATc1 signalling pathway that promotes generation of inflammatory agents in HMEC-1 cells. CONCLUSIONS: Oral treatment with Cur dampens cardiovasculopathy in acute Chagas mice. Cur impairs the Ca2+/NFATc1-regulated release of ET-1 from T. cruzi-infected vascular endothelium. These findings identify new perspectives for exploring the potential of Cur-based interventions to ameliorate Chagas heart disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chagas Cardiomyopathy/drug therapy , Curcumin/pharmacology , Endothelin-1/drug effects , NFATC Transcription Factors/drug effects , Acute Disease , Animals , Blotting, Western , Capillary Permeability/drug effects , Cells, Cultured , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/parasitology , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/parasitology , Endothelin-1/analysis , Endothelin-1/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/parasitology , Enzyme-Linked Immunosorbent Assay , Fluorescent Dyes , Interleukin-6/blood , Male , Mice, Inbred C57BL , NFATC Transcription Factors/analysis , NFATC Transcription Factors/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trypanosoma cruzi/drug effects , Tumor Necrosis Factor-alpha/blood
8.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 11-23, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28987763

ABSTRACT

Cardiac dysfunction with progressive inflammation and fibrosis is a hallmark of Chagas disease caused by persistent Trypanosoma cruzi infection. Osteopontin (OPN) is a pro-inflammatory cytokine that orchestrates mechanisms controlling cell recruitment and cardiac architecture. Our main goal was to study the role of endogenous OPN as a modulator of myocardial CCL5 chemokine and MMP-2 metalloproteinase, and its pathological impact in a murine model of Chagas heart disease. Wild-type (WT) and OPN-deficient (spp1 -/-) mice were parasite-infected (Brazil strain) for 100days. Both groups developed chronic myocarditis with similar parasite burden and survival rates. However, spp1 -/- infection showed lower heart-to-body ratio (P<0.01) as well as reduced inflammatory pathology (P<0.05), CCL5 expression (P<0.05), myocyte size (P<0.05) and fibrosis (P<0.01) in cardiac tissues. Intense OPN labeling was observed in inflammatory cells recruited to infected heart (P<0.05). Plasma concentration of MMP-2 was higher (P<0.05) in infected WT than in spp1 -/- mice. Coincidently, specific immunostaining revealed increased gelatinase expression (P<0.01) and activity (P<0.05) in the inflamed hearts from T. cruzi WT mice, but not in their spp1 -/- littermates. CCL5 and MMP-2 induction occurred preferentially (P<0.01) in WT heart-invading CD8+ T cells and was mediated via phospho-JNK MAPK signaling. Heart levels of OPN, CCL5 and MMP-2 correlated (P<0.01) with collagen accumulation in the infected WT group only. Endogenous OPN emerges as a key player in the pathogenesis of chronic Chagas heart disease, through the upregulation of myocardial CCL5/MMP-2 expression and activities resulting in pro-inflammatory and pro-hypertrophic events, cardiac remodeling and interstitial fibrosis.


Subject(s)
Atrial Remodeling , Chagas Cardiomyopathy , Chemokine CCL5/metabolism , Matrix Metalloproteinase 2/metabolism , Myocarditis , Osteopontin/physiology , Ventricular Remodeling , Animals , Atrial Remodeling/genetics , Atrial Remodeling/immunology , Cells, Cultured , Chagas Cardiomyopathy/genetics , Chagas Cardiomyopathy/immunology , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Disease Models, Animal , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Osteopontin/genetics , Ventricular Remodeling/genetics , Ventricular Remodeling/immunology
9.
Mem. Inst. Oswaldo Cruz ; 113(9): e180171, 2018. graf
Article in English | LILACS | ID: biblio-955120

ABSTRACT

BACKGROUND The anti-inflammatory and cardioprotective properties of curcumin (Cur), a natural polyphenolic flavonoid isolated from the rhizomes of Curcuma longa, are increasingly considered to have beneficial effects on the progression of Chagas heart disease, caused by the protozoan parasite Trypanosoma cruzi. OBJECTIVE To evaluate the effects of oral therapy with Cur on T. cruzi-mediated cardiovasculopathy in acutely infected mice and analyse the in vitro response of parasite-infected human microvascular endothelial cells treated with this phytochemical. METHODS Inflammation of heart vessels from Cur-treated and untreated infected mice were analysed by histology, with benznidazole (Bz) as the reference compound. Parasitaemia was monitored by the direct method. Capillary permeability was visualised by Evans-blue assay. Myocardial ET-1, IL-6, and TNF-α mRNA expressions were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Microvascular endothelial HMEC-1 cells were infected in vitro with or without addition of Cur or Bz. Induction of the Ca2+/NFAT pathway was assessed by fluorometry, immunoblotting, and reporter assay. FINDINGS Oral Cur therapy of recently infected mice reduced inflammatory cell infiltration of myocardial arteries without lowering parasite levels. Compared to that of the phosphate-buffered saline-receiving group, hearts from Cur-treated mice showed significantly decreased vessel inflammation scores (p < 0.001), vascular permeabilities (p < 0.001), and levels of IL-6/TNF-α (p < 0.01) and ET-1 (p < 0.05) mRNA. Moreover, Cur significantly (p < 0.05 for transcript; p < 0.01 for peptide) downregulated ET-1 secretion from infected HMEC-1 cells. Remarkably, Cur addition significantly (p < 0.05 at 27.0 μM) interfered with T. cruzi-dependent activation of the Ca2+/NFATc1 signalling pathway that promotes generation of inflammatory agents in HMEC-1 cells. CONCLUSIONS Oral treatment with Cur dampens cardiovasculopathy in acute Chagas mice. Cur impairs the Ca2+/NFATc1-regulated release of ET-1 from T. cruzi-infected vascular endothelium. These findings identify new perspectives for exploring the potential of Cur-based interventions to ameliorate Chagas heart disease.


Subject(s)
Humans , Chagas Cardiomyopathy/parasitology , Reverse Transcriptase Polymerase Chain Reaction , NFATC Transcription Factors/analysis , Blotting, Western , Acute Disease/rehabilitation , Anti-Inflammatory Agents/pharmacology
10.
Immunobiology ; 222(2): 423-431, 2017 02.
Article in English | MEDLINE | ID: mdl-27591076

ABSTRACT

The proinflammatory cytokine macrophage migration inhibitory factor (MIF) is a key player in innate immunity. MIF has been considered critical for controlling acute infection by the protozoan Trypanosoma cruzi, but the underlying mechanisms are poorly understood. Our study aimed to analyze whether MIF could favor microbicidal activity of the macrophage, a site where T. cruzi grows and the initial effector cell against this parasite. Using murine macrophages infected in vitro, we examined the effect of MIF on their parasiticidal ability and attempted to identify inflammatory agents involved in MIF-induced protection. Our findings show that MIF is readily secreted from peritoneal macrophages upon T. cruzi infection. MIF activates both primary and J774 phagocytes boosting the endogenous production of tumor necrosis factor-alpha via mitogen-activated protein kinase p38 signaling, as well as the release of nitric oxide and reactive oxygen species, leading to enhanced pathogen elimination. MIF can also potentiate the effect of interferon-gamma on T. cruzi killing by J774 and mouse peritoneal macrophages, rendering these cells more competent in reducing intracellular parasite burden. The present results unveil a novel innate immune pathway that contributes to host defense and broaden our understanding of the regulation of inflammatory mediators implicated in early parasite containment that is decisive for resistance to T. cruzi infection.


Subject(s)
Macrophage Activation/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/physiology , Macrophages/parasitology , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factors/metabolism , Animals , Chagas Disease/immunology , Chagas Disease/metabolism , Chagas Disease/parasitology , Female , Host-Parasite Interactions/immunology , MAP Kinase Signaling System/drug effects , Macrophage Activation/drug effects , Macrophage Migration-Inhibitory Factors/pharmacology , Macrophages/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Phagocytosis/immunology , Trypanosoma cruzi/immunology
11.
Phytomedicine ; 23(12): 1392-1400, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27765359

ABSTRACT

BACKGROUND: Diverse cardiovascular signaling routes have been considered critical for Chagas cardiomyopathy caused by the protozoan parasite Trypanosoma cruzi. Along this line, T. cruzi infection and endothelin-1 (ET-1) have been shown to cooperatively activate the Ca2+/NFAT cascade in cardiomyocytes, leading to cyclooxygenase type 2 (COX-2) induction and increased release of prostanoids and prohypertrophic peptides. PURPOSE: To determine whether the well-known cardioprotective and anti-inflammatory effects of curcumin (Cur) could be helpful to interfere with this key machinery for pathogenesis of Chagas myocarditis. STUDY DESIGN: Cur treatment was evaluated through in vivo studies using a murine model of acute T. cruzi infection and in vitro experiments using ET-1-stimulated and parasite-infected mouse cardiomyocytes. METHODS: Cur-treated and untreated infected mice were followed-up to estimate survival postinfection and heart tissues from both groups were analyzed for inflammatory infiltration by histopathology, whereas parasite load, induction of arachidonic acid pathway and natriuretic peptide expression were determined by real-time PCR. Molecular analysis of Cur myocardial targets included intracellular calcium measurement, NFAT and COX-2 induction in transfected cells, and assessment of NFAT, COX-2 and microsomal prostaglandin E synthase-1 (mPGES-1) levels by immunoblotting, prostaglandin E2 (PGE2) by ELISA, b-type natriuretic peptide (BNP) by real-time PCR, and PGE2/EP4 receptor/BNP interaction by transwell experiments. RESULTS: Cur treatment of acute Chagas mice enhanced survival and proved to hinder relevant inflammatory processes in the heart, including leukocyte recruitment, activation of the eicosanoid pathway and BNP overexpression, without modifying parasite burden in the organ. Cur was capable of blocking Ca2+-dependent NFATc1 transcriptional activity, COX-2 and mPGES-1 induction, and subsequent PGE2 production in ET-1-stimulated and parasite-infected cardiomyocytes. Furthermore, the decline of cardiomyocyte-derived prostaglandin levels achieved upon Cur treatment impaired effective PGE2/EP4 receptor interaction, resulting in attenuated expression of BNP, a strong indicator of cardiac pathogenesis in Chagas disease, in both infected and uninfected cells. CONCLUSION: Our current study shows a putative mechanism of action of Cur involving inhibition of the Ca2+/NFAT-dependent, pathogenic COX-2/mPGES-1/PGE2 pathway in T. cruzi-infected myocytes, underlying cardioprotection achieved in Cur-treated infected mice. With a view to the limited therapeutic possibilities available, Cur represents a promising approach for the treatment of Chagas heart disease.


Subject(s)
Chagas Cardiomyopathy/metabolism , Curcumin/pharmacology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , NFATC Transcription Factors/metabolism , Plant Extracts/pharmacology , Trypanosoma cruzi/pathogenicity , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Chagas Cardiomyopathy/drug therapy , Chagas Cardiomyopathy/parasitology , Chagas Disease , Curcuma/chemistry , Curcumin/therapeutic use , Cyclooxygenase 1/metabolism , Endothelin-1/metabolism , Female , Heart/drug effects , Heart/parasitology , Humans , Mice, Inbred BALB C , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phytotherapy , Plant Extracts/therapeutic use , Signal Transduction
12.
Parasitology ; 143(1): 60-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26522645

ABSTRACT

The gut epithelial barrier is a strategic place to prevent, or at least to limit, parasite dissemination upon oral infection with Toxoplasma gondii. Innate immunity to this pathogen results from delicate interactions involving different components of the infecting agent and the host. We herein aimed to examine the molecular mechanism by which protozoan DNA boosts the production of α-defensin-5 (DEFA-5), the main antimicrobial peptide at the target site of infection. The present study shows that DEFA-5 is rapidly upregulated in intestinal epithelial cells following intracellular Toll-like receptor 9 (TLR9) activation by unmethylated CpG motifs in DNA from T. gondii (CpG-DNA). Concomitantly, CpG-DNA purified from the pathogen markedly increased TLR9 mRNA expression levels in the Caco-2 cell line. We further verified that DEFA-5 production was dependent on interferon-ß released from these cells upon treatment with CpG-DNA prepared from tachyzoites. Our results suggest that, in protozoan DNA-stimulated intestinal epithelial cells, the TLR9/interferon-ß/DEFA-5 pathway may initiate an innate anti-T. gondii response without the need of parasite invasion. These findings highlight the key role of the gut epithelium in Toxoplasma recognition and amplification of local host defence against this microbe, thereby contributing to gain insight into immunoprotective mechanisms and to improve therapeutic strategies.


Subject(s)
Interferon-beta/immunology , Nucleotide Motifs/genetics , Toll-Like Receptor 9/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , alpha-Defensins/metabolism , Animals , Caco-2 Cells , DNA Methylation , DNA, Protozoan/genetics , Epithelial Cells/immunology , Humans , Immunity, Innate , Intestines/immunology , Toxoplasma/genetics , Toxoplasmosis/parasitology , alpha-Defensins/genetics , alpha-Defensins/immunology
13.
Microb Pathog ; 73: 47-52, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24927936

ABSTRACT

Trypanosoma cruzi (Tc), the etiological agent of Chagas disease, triggers multiple responses in the myocardium, a central organ of infection and pathology in the host. Parasite-driven induction of diverse regulators of cardiovascular function, including the vasoconstrictor endothelin-1 (ET-1), the inducible form of nitric oxide synthase (iNOS) and the B-type natriuretic peptide (BNP), has been linked to the development of severe chagasic cardiomyopathy. Our current goal was to analyze the participation of the zinc finger transcription factor GATA4, critically implicated in pathological cardiac hypertrophic response, in the generation of key mediators involved in the pathogenesis of Tc-elicited heart dysfunction. In this study, we found that the combined effects of Tc and ET-1 on atrial myocytes promoted the protein expression, phosphorylation and DNA-binding activity of GATA4, leading to augmented protein levels of iNOS and increased nitric oxide release. Moreover, Tc- and ET-1-co-activation of cardiomyocytes resulted in enhanced GATA4-dependent secretion of BNP. Accordingly, mice with chronic chagasic cardiomyopathy showed increased expression of GATA4, iNOS and BNP at inflammatory lesions in cardiac muscle. Our findings support a role for the GATA4 signaling pathway in the myocardial production of pathogenic mediators associated with Chagas heart disease, and may help define novel therapeutic targets.


Subject(s)
Chagas Cardiomyopathy/pathology , Endothelin-1/metabolism , GATA4 Transcription Factor/metabolism , Myocardium/pathology , Signal Transduction , Trypanosoma cruzi/pathogenicity , Animals , Cells, Cultured , Disease Models, Animal , Mice, Inbred BALB C , Natriuretic Peptide, Brain/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/biosynthesis
14.
PLoS One ; 8(2): e57181, 2013.
Article in English | MEDLINE | ID: mdl-23451183

ABSTRACT

Clinical symptoms of chronic Chagas disease occur in around 30% of the individuals infected with Trypanosoma cruzi and are characterized by heart inflammation and dysfunction. The pathogenesis of chronic chagasic cardiomyopathy (CCC) is not completely understood yet, partially because disease evolution depends on complex host-parasite interactions. Macrophage migration inhibitory factor (MIF) is a pleiotropic proinflammatory cytokine that promotes numerous pathophysiological processes. In the current study, we investigated the link between MIF and CCC progression.Immunohistochemical analysis demonstrated MIF overexpression in the hearts from chronically T. cruzi-infected mice, particularly those showing intense inflammatory infiltration. We also found that MIF exogenously added to parasite-infected murine macrophage cultures is capable of enhancing the production of TNF-α and reactive oxygen species, both with pathogenic roles in CCC. Thus, the integrated action of MIF and other cytokines and chemokines may account for leukocyte influx to the infected myocardium, accompanied by enhanced local production of multiple inflammatory mediators. We further examined by ELISA the level of MIF in the sera from chronic indeterminate and cardiomyopathic chagasic patients, and healthy subjects. CCC patients displayed significantly higher MIF concentrations than those recorded in asymptomatic T. cruzi-infected and uninfected individuals. Interestingly, increased MIF levels were associated with severe progressive Chagas heart disease, in correlation with elevated serum concentration of high sensitivity C-reactive protein and also with several echocardiographic indicators of left ventricular dysfunction, one of the hallmarks of CCC. Our present findings represent the first evidence that enhanced MIF production is associated with progressive cardiac impairment in chronic human infection with T. cruzi, strengthening the relationship between inflammatory response and parasite-driven pathology. These observations contribute to unravel the elements involved in the pathogenesis of CCC and may also be helpful for the design of novel therapies aimed to control long-term morbidity in chagasic patients.


Subject(s)
Cardiomyopathies/blood , Chagas Disease/blood , Macrophage Migration-Inhibitory Factors/blood , Animals , Cardiomyopathies/complications , Cardiomyopathies/pathology , Cell Line , Chagas Disease/complications , Chronic Disease , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Macrophages/metabolism , Mice , Mice, Inbred C3H , Middle Aged , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
PLoS Negl Trop Dis ; 7(2): e2034, 2013.
Article in English | MEDLINE | ID: mdl-23409199

ABSTRACT

Trypanosoma cruzi, the causative agent of Chagas' disease, induces multiple responses in the heart, a critical organ of infection and pathology in the host. Among diverse factors, eicosanoids and the vasoactive peptide endothelin-1 (ET-1) have been implicated in the pathogenesis of chronic chagasic cardiomyopathy. In the present study, we found that T. cruzi infection in mice induces myocardial gene expression of cyclooxygenase-2 (Cox2) and thromboxane synthase (Tbxas1) as well as endothelin-1 (Edn1) and atrial natriuretic peptide (Nppa). T. cruzi infection and ET-1 cooperatively activated the Ca(2+)/calcineurin (Cn)/nuclear factor of activated T cells (NFAT) signaling pathway in atrial myocytes, leading to COX-2 protein expression and increased eicosanoid (prostaglandins E(2) and F(2α), thromboxane A(2)) release. Moreover, T. cruzi infection of ET-1-stimulated cardiomyocytes resulted in significantly enhanced production of atrial natriuretic peptide (ANP), a prognostic marker for impairment in cardiac function of chagasic patients. Our findings support an important role for the Ca(2+)/Cn/NFAT cascade in T. cruzi-mediated myocardial production of inflammatory mediators and may help define novel therapeutic targets.


Subject(s)
Endothelin-1/biosynthesis , Myocytes, Cardiac/immunology , Myocytes, Cardiac/parasitology , Trypanosoma cruzi/immunology , Trypanosoma cruzi/pathogenicity , Animals , Atrial Natriuretic Factor , Calcineurin/metabolism , Calcium/metabolism , Cyclooxygenase 2/biosynthesis , Gene Expression Profiling , Inflammation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/parasitology , NFATC Transcription Factors/metabolism , Natriuretic Peptide, C-Type/biosynthesis , Protein Precursors/biosynthesis , Signal Transduction , Thromboxane-A Synthase/biosynthesis
16.
Hum Vaccin Immunother ; 9(2): 409-12, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23291939

ABSTRACT

Archaeosomes (ARC), vesicles made from lipids extracted from Archaea, display strong adjuvant properties. In this study, we evaluated the ability of the highly stable ARC formulated from total polar lipids of a new Halorubrum tebenquichense strain found in Argentinean Patagonia, to act as adjuvant for soluble parasite antigens in developing prophylactic vaccine against the intracellular protozoan T. cruzi, the etiologic agent of Chagas disease. We demonstrated for the first time that C3H/HeN mice subcutaneously immunized with trypanosomal antigens entrapped in these ARC (ARC-TcAg) rapidly developed higher levels of circulating T. cruzi antibodies than those measured in the sera from animals receiving the antigen alone. Enhanced humoral responses elicited by ARC-TcAg presented a dominant IgG2a antibody isotype, usually associated with Th1-type immunity and resistance against T. cruzi. More importantly, ARC-TcAg-vaccinated mice displayed reduced parasitemia during early infection and were protected against an otherwise lethal challenge with the virulent Tulahuén strain of the parasite. Our findings suggest that, as an adjuvant, H. tebenquichense-derived ARC may hold great potential to develop a safe and helpful vaccine against this relevant human pathogen.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Chagas Disease/prevention & control , Halorubrum/chemistry , Liposomes/administration & dosage , Membrane Lipids/administration & dosage , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/immunology , Adjuvants, Immunologic/isolation & purification , Animals , Antibodies, Protozoan/blood , Argentina , Chagas Disease/immunology , Disease Models, Animal , Female , Immunoglobulin G/blood , Injections, Subcutaneous , Liposomes/isolation & purification , Membrane Lipids/isolation & purification , Mice, Inbred C3H , Parasitemia/immunology , Parasitemia/prevention & control , Survival Analysis , Th1 Cells/immunology , Treatment Outcome
17.
Cytokine ; 61(2): 491-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23199812

ABSTRACT

Osteopontin (OPN) is a multifunctional protein participating in the regulation of different Th cell lineages and critically involved in the initiation of immune responses to diverse pathogens. Our study goal was to verify whether OPN helps modulate the protective Th1 and Th17 cytokine responses in C57BL/6 mice infected with Trypanosoma cruzi, the etiological agent of Chagas disease. Parasite infection induced OPN release from murine macrophages in vitro and acute Chagas mice displayed enhanced serum levels of this cytokine at the peak of parasitemia. Upon administration of a neutralizing anti-OPN antibody, recently infected mice presented lower Th1 and Th17 responses, increased parasitemia and succumbed earlier and at higher rates to infection than non-immune IgG-receiving controls. The anti-OPN therapy also resulted in reduced circulating levels of IL-12 p70, IFN-γ, IL-17A and specific IgG(2a) antibodies. Furthermore, antibody-mediated blockade of OPN activity abrogated the ex vivo production of IL-12 p70, IFN-γ and IL-17A, while promoting IL-10 secretion, by spleen macrophages and CD4(+) T cells from T. cruzi-infected mice. Th1 and Th17 cytokine release induced by OPN preferentially involved the α(v)ß(3) integrin OPN receptor, whereas concomitant down-modulation of IL-10 production would mostly depend on OPN interaction with CD44. Our findings suggest that, in resistant C57BL/6 mice, elicitation of protective Th1 and Th17 cytokine responses to T. cruzi infection is likely to be regulated by endogenous OPN.


Subject(s)
Chagas Disease/immunology , Chagas Disease/parasitology , Osteopontin/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Trypanosoma cruzi/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/parasitology , Bone Marrow Cells/pathology , Cell Adhesion , Chagas Disease/blood , Disease Progression , Female , Host-Parasite Interactions/immunology , Immunity/immunology , Inflammation Mediators/metabolism , Interleukin-12/biosynthesis , Interleukin-12/blood , Lymphocyte Count , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/parasitology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Osteopontin/biosynthesis , Parasitemia/immunology , Spleen/immunology , Spleen/parasitology , Spleen/pathology , Survival Analysis
18.
PLoS One ; 6(10): e27133, 2011.
Article in English | MEDLINE | ID: mdl-22066031

ABSTRACT

OBJECTIVE: The presence of autoantibodies with adrenergic and cholinergic activity, capable of triggering neurotransmitter receptor-mediated effects, has been associated with pathogenesis in T. cruzi-infected hosts. The goal of this study was to investigate the production of anti-M2 muscarinic receptor autoantibodies (Anti-M2R AAbs) as well as the IFN-γ profile in children at the early stage of Chagas disease, and to examine whether trypanocidal chemotherapy with benzonidazole (BZ) could modify both response patterns. METHODS: This study comprised 30 T. cruzi-infected children (mean age: 13.8 years) and 19 uninfected controls (mean age: 12.7 years). Infected patients were treated with BZ and followed-up. Blood samples collected at diagnosis-T0, end of treatment-T1, and six months later-T2 were analysed by ELISA for detection of Anti-M2R AAbs and circulating levels of IFN-γ. RESULTS: At T0, anti-M2R AAbs were demonstrated in 56.7% of T. cruzi-infected patients, whereas uninfected controls were 100% negative. The average age of Anti-M2R AAbs(+) patients was higher than that from negative population. Infected children also displayed significantly stronger serum IFN-γ responses than controls. Upon BZ treatment, a significant linear decreasing trend in Anti-M2R AAb reactivity was recorded throughout the follow-up, with 29.7-88.1% decrease at T2. IFN-γ circulating levels also declined by T2. CONCLUSION: Anti-M2R AAbs and IFN-γ raise early during chagasic infection in children and are downmodulated by BZ therapy. These findings reinforce the usefulness of early BZ treatment not only to eliminate the parasite but also to reduce potentially pathogenic immune responses.


Subject(s)
Autoantibodies/immunology , Chagas Disease/drug therapy , Chagas Disease/parasitology , Interferon-gamma/immunology , Nitroimidazoles/therapeutic use , Receptor, Muscarinic M2/immunology , Trypanosoma cruzi/immunology , Adolescent , Autoantibodies/blood , Case-Control Studies , Chagas Disease/blood , Chagas Disease/immunology , Child , Female , Humans , Male , Treatment Outcome
19.
Exp Parasitol ; 128(4): 448-53, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21554876

ABSTRACT

Synthetic oligodeoxynucleotides containing unmethylated CpG motifs (CpG-ODN) have been characterized as Th1-promoting immunopotentiators, an adjuvant activity desirable for vaccination against intracellular parasites like Toxoplasma gondii. In an attempt to find new antigen-adjuvant combinations that enhance the immunogenicity of antigen candidates for toxoplasma vaccines, we analyzed the extent of protection in mice immunized with ROP2 and GRA4 recombinant proteins when co-administered with CpG-ODN. Both GRA4+CpG-ODN and ROP2+CpG-ODN formulations were shown to induce a strong humoral Th1-biased response characterized by a high IgG(2a) to IgG(1) antibody ratio. Both vaccination regimens led to increased secretion of IFN-γ and IL-10, and negligible amounts of IL-4, upon specific re-stimulation of spleen cells from these groups of mice. After a non-lethal challenge with tissue cysts of a moderately virulent strain, only the brains from mice vaccinated with ROP2 or GRA4 in combination with CpG-ODN showed a significant reduction (63% and 62%, respectively) in their parasite load compared to the controls. The rate of protection obtained with GRA4+ROP2+CpG-ODN resulted equivalent (66%) to those achieved with the single antigens plus CpG-ODN. Taken together, these results indicate that CpG-ODN is an important candidate adjuvant for use in potential multicomponent anti-T. gondii vaccines for animals and humans.


Subject(s)
Membrane Proteins/immunology , Oligodeoxyribonucleotides/immunology , Protozoan Proteins/immunology , Recombinant Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/prevention & control , Adjuvants, Immunologic , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/blood , Brain/parasitology , Cytokines/analysis , Female , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Protozoan Proteins/genetics , Protozoan Vaccines/immunology , Spleen/immunology , Toxoplasmosis, Animal/immunology
20.
J Infect Dis ; 202(1): 136-44, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20497050

ABSTRACT

Despite the strong immune responses elicited after natural infection with Trypanosoma cruzi or vaccination against it, parasite survival suggests that these responses are insufficient or inherently inadequate. T. cruzi contains a major cystein proteinase, cruzipain, which has a catalytic N-terminal domain and a C-terminal extension. Immunizations that employed recombinant cruzipain or its N- and C-terminal domains allowed evaluation of the ability of cruzipain to circumvent responses against the catalytic domain. This phenomenon is not a property of the parasite but of cruzipain itself, because recombinant cruzipain triggers a response similar to that of cruzipain during natural or experimental infection. Cruzipain is not the only antigen with a highly immunogenic region of unknown function that somehow protects an essential domain for parasite survival. However, our studies show that this can be reverted by using the N-terminal domain as a tailored immunogen able to redirect host responses to provide enhanced protection.


Subject(s)
Chagas Disease/prevention & control , Cysteine Endopeptidases/immunology , Protozoan Vaccines/immunology , Trypanosoma cruzi/genetics , Animals , Antibodies, Protozoan/blood , Female , Mice , Mice, Inbred C3H , Muscle, Skeletal/pathology , Myocardium/pathology , Protozoan Proteins/immunology , Recombinant Proteins/immunology , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...