Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
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.
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
7.
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
8.
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
9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...