Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Immunology ; 168(3): 444-458, 2023 03.
Article En | MEDLINE | ID: mdl-36164989

Arthralgia is a hallmark of chikungunya virus (CHIKV) infection and can be very debilitating and associated with a robust local inflammatory response. Many pathophysiological aspects associated with the disease remain to be elucidated. Here, we describe a novel model of CHIKV infection in immunocompetent mice and evaluate the role of tumour necrosis factor in the pathogenesis of the disease. C57BL/6 wild type (WT) or TNF receptor 1 deficient (TNFR1-/- ) mice were inoculated with 1 × 106 PFU of CHIKV in the paw. Alternatively, etanercept was used to inhibit TNF in infected WT mice. Hypernociception, inflammatory and virological analysis were performed. Inoculation of CHIKV into WT mice induced persistent hypernociception. There was significant viral replication in target organs and local production of inflammatory mediators in early time-points after infection. CHIKV infection was associated with specific humoral IgM and IgG responses. In TNFR1-/- mice, there was a decrease in the hypernociception threshold, which was associated with a milder local inflammatory response in the paw but delayed viral clearance. Local or systemic treatment with etanercept reduced CHIKV-induced hypernociception. This is the first study to describe hypernociception, a clinical correlation of arthralgia, in immunocompetent mice infected with CHIKV. It also demonstrates the dual role of TNF in contributing to viral clearance but driving tissue damage and hypernociception. Inhibition of TNF may have therapeutic benefits but its role in viral clearance suggests that viral levels must be monitored in CHIKV-infected patients and that TNF inhibitors should ideally be used in combination with anti-viral drugs.


Chikungunya Fever , Chikungunya virus , Animals , Mice , Chikungunya Fever/pathology , Receptors, Tumor Necrosis Factor, Type I , Etanercept , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha , Virus Replication , Arthralgia
2.
Clin Immunol ; 234: 108913, 2022 01.
Article En | MEDLINE | ID: mdl-34954347

Chagas disease has a complex pathogenesis wherein the host immune response is essential for controlling its development. Suppressor of cytokine signaling(SOCS)2 is a crucial protein that regulates cytokine production. In this study, SOCS2 deficiency resulted in an initial imbalance of IL12- and IL-10-producing neutrophils and dendritic cells (DCs), which caused a long-lasting impact reducing inflammatory neutrophils and DCs, and tolerogenic DCs at the peak of acute disease. A reduced number of inflammatory and pro-resolving macrophages, and IL17A-producing CD4+ T cells, and increased lymphocyte apoptosis was found in SOCS2-deficient mice. Electrocardiogram analysis of chimeric mice showed that WT mice that received SOCS2 KO bone marrow transplantation presented increased heart dysfunction. Taken together, the results demonstrated that SOCS2 is a crucial regulator of the immune response during Trypanosoma cruzi infection, and suggest that a SOCS2 genetic polymorphism, or failure of its expression, may increase the susceptibility of cardiomyopathy development in Chagasic patients.


Cardiomyopathies/etiology , Chagas Disease/immunology , Dendritic Cells/immunology , Neutrophils/immunology , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Bone Marrow Transplantation , Chagas Disease/complications , Female , Mice , Mice, Inbred C57BL , Spleen/immunology , Suppressor of Cytokine Signaling Proteins/genetics , Th17 Cells/immunology
3.
Clin Immunol ; 226: 108713, 2021 05.
Article En | MEDLINE | ID: mdl-33711450

Current chemical therapies for Chagas Disease (CD) lack ability to clear Trypanosoma cruzi (Tc) parasites and cause severe side effects, making search for new strategies extremely necessary. We evaluated the action of Tityus serrulatus venom (TsV) components during Tc infection. TsV treatment increased nitric oxide and pro-inflammatory cytokine production by Tc-infected macrophages (MØ), decreased intracellular parasite replication and trypomastigotes release, also triggering ERK1/2, JNK1/2 and p38 activation. Ts7 demonstrated the highest anti-Tc activity, inducing high levels of TNF and IL-6 in infected MØ. TsV/Ts7 presented synergistic effect on p38 activation when incubated with Tc antigen. KPP-treatment of MØ also decreased trypomastigotes releasing, partially due to p38 activation. TsV/Ts7-pre-incubation of Tc demonstrated a direct effect on parasite decreasing MØ-trypomastigotes releasing. In vivo KPP-treatment of Tc-infected mice resulted in decreased parasitemia. Summarizing, this study opens perspectives for new bioactive molecules as CD-therapeutic treatment, demonstrating the TsV/Ts7/KPP-trypanocidal and immunomodulatory activity during Tc infection.


Chagas Disease/drug therapy , Immunomodulation/drug effects , Scorpion Venoms/pharmacology , Scorpions/metabolism , Animals , Chagas Disease/metabolism , Female , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Tumor Necrosis Factors/metabolism
4.
Mediators Inflamm ; 2019: 1872593, 2019.
Article En | MEDLINE | ID: mdl-31949423

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS). Experimental Autoimmune Encephalomyelitis (EAE) is the most widely used animal model for the study of MS. The Suppressor of Cytokine Signaling (SOCS) 2 protein plays a critical role in regulating the immune responses. The role of SOCS2 during EAE has not been explored. EAE was induced in WT and SOCS2-/- mice using myelin oligodendrocyte glycoprotein (MOG35-55) peptide. Brain and spinal cord were examined during the peak (day 14) and recovery phase (day 28) of the disease. SOCS2 was upregulated in the brain of WT mice at the peak and recovery phase of EAE. The development of the acute phase was slower in onset in SOCS2-/- mice and was associated with reduced number of Th1 (CD3+CD4+IFN-γ +) cells in the spinal cord and brain. However, while in WT mice, maximal clinical EAE score was followed by a progressive recovery; the SOCS2-/- mice were unable to recover from locomotor impairment that occurred during the acute phase. There was a prolonged inflammatory response (increased Th1 and decreased Th2 and T regulatory cells) in the late phase of EAE in the CNS of SOCS2-/- mice. Transplantation of bone marrow cells from SOCS2-/- into irradiated WT mice resulted in higher lethality at the early phase of EAE. Altogether, these results suggest that SOCS2 plays a dual role in the immune response during EAE. It is necessary for damage during the acute phase damage but plays a beneficial role in the recovery stage of the disease.


Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Blotting, Western , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Flow Cytometry , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling Proteins/genetics , Th1 Cells/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism
5.
J Exp Med ; 214(11): 3399-3415, 2017 Nov 06.
Article En | MEDLINE | ID: mdl-28947611

Leukotriene B4 (LTB4), a proinflammatory mediator produced by the enzyme 5-lipoxygenase (5-LO), is associated with the development of many inflammatory diseases. In this study, we evaluated the participation of the 5-LO/LTB4 axis in graft-versus-host disease (GVHD) pathogenesis by transplanting 5-LO-deficient leukocytes and investigated the effect of pharmacologic 5-LO inhibition by zileuton and LTB4 inhibition by CP-105,696. Mice that received allogeneic transplant showed an increase in nuclear 5-LO expression in splenocytes, indicating enzyme activation after GVHD. Mice receiving 5-LO-deficient cell transplant or zileuton treatment had prolonged survival, reduced GVHD clinical scores, reduced intestinal and liver injury, and decreased levels of serum and hepatic LTB4 These results were associated with inhibition of leukocyte recruitment and decreased production of cytokines and chemokines. Treatment with CP-105,696 achieved similar effects. The chimerism or the beneficial graft-versus-leukemia response remained unaffected. Our data provide evidence that the 5-LO/LTB4 axis orchestrates GVHD development and suggest it could be a target for the development of novel therapeutic strategies for GVHD treatment.


Arachidonate 5-Lipoxygenase/metabolism , Cell Transplantation/methods , Graft vs Host Disease/metabolism , Leukotriene B4/metabolism , Animals , Arachidonate 5-Lipoxygenase/genetics , Benzopyrans/pharmacology , Carboxylic Acids/pharmacology , Cell Transplantation/adverse effects , Chemokines/metabolism , Cytokines/metabolism , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Leukocytes/cytology , Leukocytes/enzymology , Leukocytes/metabolism , Leukotriene Antagonists/pharmacology , Leukotriene B4/antagonists & inhibitors , Lipoxygenase Inhibitors/pharmacology , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Confocal , Transplantation, Homologous
6.
Infect Immun ; 84(10): 3071-82, 2016 10.
Article En | MEDLINE | ID: mdl-27481250

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor involved in controlling several aspects of immune responses, including the activation and differentiation of specific T cell subsets and antigen-presenting cells, thought to be relevant in the context of experimental Trypanosoma cruzi infection. The relevance of AhR for the outcome of T. cruzi infection is not known and was investigated here. We infected wild-type (WT) mice and AhR knockout (AhR KO) mice with T. cruzi (Y strain) and determined levels of parasitemia, myocardial inflammation and fibrosis, expression of AhR/cytokines/suppressor of cytokine signaling (SOCS) (spleen/heart), and production of nitric oxide (NO), reactive oxygen species (ROS), and peroxynitrite (ONOO(-)) (spleen). AhR expression was increased in the heart of infected WT mice. Infected AhR KO mice displayed significantly reduced parasitemia, inflammation, and fibrosis of the myocardium. This was associated with an anticipated increased immune response characterized by increased levels of inflammatory cytokines and reduced expression of SOCS2 and SOCS3 in the heart. In vitro, AhR deficiency caused impairment in parasite replication and decreased levels of ROS production. In conclusion, AhR influences the development of murine Chagas disease by modulating ROS production and regulating the expression of key physiological regulators of inflammation, SOCS1 to -3, associated with the production of cytokines during experimental T. cruzi infection.


Chagas Disease/physiopathology , Cytokines/metabolism , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/physiology , Trypanosoma cruzi/physiology , Animals , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Chagas Disease/metabolism , Chagas Disease/pathology , Disease Models, Animal , Mice , Mice, Knockout , Myocarditis/metabolism , Myocarditis/pathology , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Spleen/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism
7.
Brain Behav Immun ; 54: 73-85, 2016 May.
Article En | MEDLINE | ID: mdl-26765997

Plasmodium falciparum infection results in severe malaria in humans, affecting various organs, including the liver, spleen and brain, and resulting in high morbidity and mortality. The Plasmodium berghei ANKA (PbA) infection in mice closely recapitulates many aspects of human cerebral malaria (CM); thus, this model has been used to investigate the pathogenesis of CM. Suppressor of cytokine signaling 2 (SOCS2), an intracellular protein induced by cytokines and hormones, modulates the immune response, neural development, neurogenesis and neurotrophic pathways. However, the role of SOCS2 during CM remains unknown. SOCS2 knockout (SOCS2(-/-)) mice infected with PbA show an initial resistance to infection with reduced parasitemia and production of TNF, TGF-ß, IL-12 and IL-17 in the brain. Interestingly, in the late phase of infection, SOCS2(-/-) mice display increased parasitemia and reduced Treg cell infiltration, associated with enhanced levels of Th1 and Th17 cells and related cytokines IL-17, IL-6, and TGF-ß in the brain. A significant reduction in protective neurotrophic factors, such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), was also observed. Moreover, the molecular alterations in the brain of infected SOCS2(-/-) mice were associated with anxiety-related behaviors and cognition impairment. Mechanistically, these results revealed enhanced nitric oxide (NO) production in PbA-infected SOCS2(-/-) mice, and the inhibition of NO synthesis through l-NAME led to a marked decrease in survival, the disruption of parasitemia control and more pronounced anxiety-like behavior. Treatment with l-NAME also shifted the levels of Th1, Th7 and Treg cells in the brains of infected SOCS2(-/-) mice to the background levels observed in infected WT, with remarkable exception of increased CD8(+)IFN(+) T cells and inflammatory monocytes. These results indicate that SOCS2 plays a dual role during PbA infection, being detrimental in the control of the parasite replication but crucial in the regulation of the immune response and production of neurotrophic factors. Here, we provided strong evidence of a critical relationship between SOCS2 and NO in the orchestration of the immune response and development of CM during PbA infection.


Malaria, Cerebral/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Malaria, Cerebral/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/metabolism , Plasmodium berghei/isolation & purification , Spleen/metabolism , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Transforming Growth Factor beta/metabolism
8.
Belo Horizonte; s.n; 2016. 101 p. ilus, graf.
Thesis Pt | LILACS, ColecionaSUS | ID: biblio-1427245

A infecção por Trypanosoma cruzi (T. cruzi), causador da doença de Chagas, induz uma reação inflamatória e a eficiência da resposta imune (RI) do hospedeiro é importante para que a infecção persista ou seja eliminada. A expressão de SOCS2 (Supressor de Sinalização de Citocinas)2, uma proteína intracelular, é parcialmente mediada por lipoxinas (LXA4, eicosanoide anti-inflamatório) em células dendríticas (DCs), a principal célula apresentadora de antígeno (APC). Demonstramos que SOCS2 é fundamental durante a infecção por T. cruzi modulando a geração/expansão de células Th1, Treg e de memória e no controle da função cardíaca. No presente trabalho, nós pesquisamos o papel de SOCS2 na função de DCs e na indução/manutenção da RI durante a infecção experimental por T. cruzi. Camundongos CD11cDTR transgênicos (depleção de DCs), selvagens (WT) e deficientes de SOCS2 (knockout/KO) foram infectados com a cepa Y de T. cruzi. Nossos resultados demonstraram um aumento da parasitemia nos animais depletados de DCs, ressaltando que DCs são cruciais no controle da infecção por T. cruzi. Durante a RI inata a ausência de SOCS2 resultou na redução da frequência de DCs produtoras de citocinas inflamatórias (IL-12 e TNF-α), mas não de IL-10, sem alterar a expressão dos receptores do tipo Toll (TLR2 e TLR4) e de MHC II. Em contraste, uma diminuição na expressão da molécula co-estimuladora CD80 foi observada em DCs deficientes de SOCS2. Durante a RI adaptativa a ausência de SOCS2 em DCs resultou no aumento dos níveis de expressão de TLR2 e TLR4 e na redução da frequência de DCs expressando MHCII. A transferência adotiva de DCs deficientes de SOCS2 ocasionou aumento da parasitemia e mudanças do perfil da RI frente a infecção por T. cruzi, principalmente: i) reduzindo a frequência de células NK produtoras de IFN-γ e de IL17; ii) diminuindo a frequência de células T CD8 produtoras de IFN-γ e de T CD4 produtoras de IL-17, apesar de aumentar células T CD4 produtoras de IFN-γ; ausência de SOCS2 em DCs também resultou em redução de células produtoras de IL-10, como T CD4 e CD19, além das células Treg. Nossos resultados também demonstraram que SOCS2 é importante na modulação da apoptose durante a infecção por T. cruzi, onde a deficiência de SOCS2 leva a um aumento da apoptose de neutrófilos durante a RI inata, de macrófagos nas RI inata e adaptativa e de linfócitos na RI adaptativa. Nossos resultados in vitro demonstraram um aumento de caspase 3 total e clivada em neutrófilos deficientes em SOCS2. Em conjunto, nossos resultados demonstraram que SOCS2 é crucial na modulação das funções de DCs durante a geração e regulação da RI inata e adaptativa durante a infecção por T. cruzi.


The infection by Trypanosoma cruzi (T. cruzi), which causes Chagas' disease, induces an inflammatory reaction and the efficacy of the host immune response (IR) is important to persist or eliminate the infection. SOCS2 (supressor of cytokine signaling)2 expression, an intracellular protein, is partially mediated by lipoxins (LXA4, anti-inflammatory eicosanoid) in dendritic cells (DCs), the main antigen-presenting cell (APC). We demonstrated that SOCS2 is fundamental during T. cruzi infection by modulating the generation/expansion of Th1, Treg and memory cells and in the control of heart function. In the present work, we investigated the role of SOCS2 in DCs function and induction/maintenance of IR during experimental T. cruzi infection. CD11cDTR transgenic mice (DCs depletion), wild type (WT) and SOCS2 (knockout/KO) were infected with Y strain of T. cruzi. Our results demonstrated an increased parasitemia in depleted DCs animals, emphasizing that DCs are crucial in control of T. cruzi infection. During innate IR, absence of SOCS2 resulted in decreased frequency of inflammatory cytokines (IL-12 and TNF-α), but not IL-10 by DCs, without change the Tolllike receptor expression (TLR2 and TLR4) and MHCII. In contrast, a decreased expression of CD80 costimulatory molecule was observed in SOCS2 deficient DCs. During adaptive IR, absence of SOCS2 in DCs resulted in increased levels of TLR2 and TLR4 expression and reduced frequency of DCs expressing MHCII. Adoptive transfer of SOCS2 deficient DCs caused increased parasitemia and changes in IR profile against T. cruzi infection, specifically: i) reducing the frequency of NK cells producing IFN-γ and IL-17; ii) reducing the frequency of CD8 T cells producing IFN-γ and CD4 T cells producing IL-17, despite increasing CD4 T cells producing IFN-γ; absence of SOCS2 in DCs also resulted in reduction of cells producing IL-10 such as CD4 and CD19, besides Treg cells. Our results also demonstrated that SOCS2 is important in apoptosis modulation during T. cruzi infection, where absence of SOCS2 leads to increased apoptosis in neutrophils during innate IR, macrophages in innate and adaptive IR and lymphocytes in adaptive IR. Our in vitro results demonstrated an increase in cleaved and total caspase 3 in SOCS2 deficient neutrophils. Together, our results demonstrated that SOCS2 is crucial in the modulation of DCs' function during generation/regulation of innate and adaptive IR during T. cruzi infection


Trypanosoma cruzi , Dendritic Cells , Chagas Cardiomyopathy , Chagas Disease , Academic Dissertation
9.
Malar J ; 14: 311, 2015 Aug 11.
Article En | MEDLINE | ID: mdl-26260055

BACKGROUND: Cerebral malaria (CM) is debilitating and sometimes fatal. Disease severity has been associated with poor treatment access, therapeutic complexity and drug resistance and, thus, alternative therapies are increasingly necessary. In this study, the effect of the administration of Agaricus blazei, a mushroom of Brazilian origin in a model of CM caused by Plasmodium berghei, strain ANKA, was investigated in mice. METHODS: C57BL/6 mice were pre-treated with aqueous extract or fractions of A. blazei, or chloroquine, infected with P. berghei ANKA and then followed by daily administration of A. blazei or chloroquine. Parasitaemia, body weight, survival and clinical signs of the disease were evaluated periodically. The concentration of pro-and anti-inflammatory cytokines, histopathology and in vitro analyses were performed. RESULTS: Mice treated with A. blazei aqueous extract or fraction C, that shows antioxidant activity, displayed lower parasitaemia, increased survival, reduced weight loss and protection against the development of CM. The administration of A. blazei resulted in reduced levels of TNF, IL-1ß and IL-6 production when compared to untreated P. berghei-infected mice. Agaricus blazei (aqueous extract or fraction C) treated infected mice displayed reduction of brain lesions. Although chloroquine treatment reduced parasitaemia, there was increased production of proinflammatory cytokines and damage in the CNS not observed with A. blazei treatment. Moreover, the in vitro pretreatment of infected erythrocytes followed by in vivo infection resulted in lower parasitaemia, increased survival, and little evidence of clinical signs of disease. CONCLUSIONS: This study strongly suggests that the administration of A. blazei (aqueous extract or fraction C) was effective in improving the consequences of CM in mice and may provide novel therapeutic strategies.


Agaricus/chemistry , Anti-Inflammatory Agents/pharmacology , Antimalarials/pharmacology , Biological Products/pharmacology , Malaria, Cerebral/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Antimalarials/chemistry , Antimalarials/therapeutic use , Biological Products/chemistry , Biological Products/therapeutic use , Brain/drug effects , Brain/pathology , Cytokines/blood , Female , Malaria, Cerebral/physiopathology , Malaria, Cerebral/prevention & control , Mice , Mice, Inbred C57BL
10.
Curr Opin Infect Dis ; 28(3): 246-52, 2015 Jun.
Article En | MEDLINE | ID: mdl-25887609

PURPOSE OF REVIEW: American trypanosomiasis, or Chagas disease, is a lifelong and persistent infection caused by the protozoan Trypanosoma cruzi and is the most significant cause of morbidity and mortality in South and Central America. Owing to immigration and additional risks from blood transfusion and organ transplantation, the number of reported cases of Chagas disease has increased recently in Europe and the USA. The disease is caused by a moderate to intense lasting inflammatory response that triggers local expression of inflammatory mediators and activates and recruits leukocytes to various tissues to eliminate the parasites. RECENT FINDINGS: This long-term inflammatory process triggers biochemical, physiological and morphological alterations and clinical changes in the digestive, nervous and cardiac (e.g. myocarditis, arrhythmias, congestive heart failure, autonomic dysfunctions and microcirculatory disturbances) systems. Indeed, the pathogenesis of Chagas disease is intricate and multifactorial, and the roles of the parasite and the immune response in initiating and maintaining the disease are still controversial. SUMMARY: In this review, we discuss the current knowledge of 'strategies' employed by the parasite to persist in the host and host defence mechanisms against Trypanosoma cruzi infection, which can result in equilibrium (absence of the disease) or disease development, mainly in the cardiac systems.


Chagas Disease/physiopathology , Inflammation/parasitology , Myocardium/pathology , Trypanocidal Agents/therapeutic use , Trypanosoma cruzi/genetics , Antibodies, Protozoan/blood , Autoimmunity , Chagas Disease/immunology , Chagas Disease/parasitology , Chagas Disease/pathology , Heart/parasitology , Host-Parasite Interactions , Humans , Trypanosoma cruzi/isolation & purification
11.
Infect Immun ; 82(8): 3127-40, 2014 Aug.
Article En | MEDLINE | ID: mdl-24818665

Infection with Plasmodium falciparum may result in severe disease affecting various organs, including liver, spleen, and brain, resulting in high morbidity and mortality. Plasmodium berghei Anka infection of mice recapitulates many features of severe human malaria. The aryl hydrocarbon receptor (AhR) is an intracellular receptor activated by ligands important in the modulation of the inflammatory response. We found that AhR-knockout (KO) mice infected with P. berghei Anka displayed increased parasitemia, earlier mortality, enhanced leukocyte-endothelial cell interactions in the brain microvasculature, and increased inflammation in brain (interleukin-17 [IL-17] and IL-6) and liver (gamma interferon [IFN-γ] and tumor necrosis factor alpha [TNF-α]) compared to infected wild-type (WT) mice. Infected AhR-KO mice also displayed a reduction in cytokines required for host resistance, including TNF-α, IL-1ß, and IFN-γ, in the brain and spleen. Infection of AhR-KO mice resulted in an increase in T regulatory cells and transforming growth factor ß, IL-6, and IL-17 in the brain. AhR modulated the basal expression of SOCS3 in spleen and brain, and P. berghei Anka infection resulted in enhanced expression of SOCS3 in brain, which was absent in infected AhR-KO mice. These data suggest that AhR-mediated control of SOCS3 expression is probably involved in the phenotype seen in infected AhR-KO mice. This is, to our knowledge, the first demonstration of a role for AhR in the pathogenesis of malaria.


Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Malaria/immunology , Malaria/pathology , Plasmodium berghei/immunology , Receptors, Aryl Hydrocarbon/immunology , Receptors, Aryl Hydrocarbon/metabolism , Suppressor of Cytokine Signaling Proteins/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/immunology , Brain/pathology , Cytokines/immunology , Cytokines/metabolism , Gene Deletion , Humans , Liver/immunology , Liver/pathology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Aryl Hydrocarbon/genetics , Spleen/immunology , Suppressor of Cytokine Signaling 3 Protein
12.
Microbes Infect ; 16(6): 481-90, 2014 Jun.
Article En | MEDLINE | ID: mdl-24704475

Chagas disease, caused by Trypanosoma cruzi (Tc), is an important cause of heart disease. Resistance to Tc infection is multifactorial and associated with Th1 response. IL-18 plays an important role in regulation of IFN-γ production/development of Th1 response. However, the role of IL-18 in the setting of Tc infection remains unclear. Therefore, we investigated the role of IL-18 in the modulation of immune response and myocarditis in Tc infection. C57BL/6 and IL-18 KO mice were infected with Tc (Y or Colombian strain) and parasitemia, immune response and pathology were evaluated. Y strain infection of IL-18 KO did not alter any parameters when compared with C57BL/6 mice. However, during the acute phase (20 and 40 days post infection-dpi), Colombian strain infected-IL-18 KO mice displayed higher serum levels of IL-12 and IFN-γ, respectively, and at the chronic phase (100 dpi) an increase in splenic IFN-γ-producing CD4(+) and CD8(+) T memory cells. There was an IL-10, FOXP3 and CD4(+)CD25(+) cells reduction during acute infection in spleen. Additionally, there was a significant reduction in leukocyte infiltration and parasite load in myocardium of chronically infected IL-18 KO mice. Collectively, these data indicate that IL-18 contributes to the pathogenesis of Tc-induced myocarditis when infected with Colombian but not Y strain. These observations also underscore that parasite and host strain differences are important in evaluation of experimental Tc infection pathogenesis.


Chagas Disease/immunology , Interleukin-18/immunology , Myocarditis/immunology , Trypanosoma cruzi/pathogenicity , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Chagas Disease/complications , Host-Parasite Interactions , Interferon-gamma/blood , Interleukin-12/blood , Interleukin-18/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/complications , Myocarditis/parasitology , Myocardium/pathology , Spleen/cytology , Spleen/immunology , Spleen/parasitology
13.
Semin Immunopathol ; 34(6): 753-70, 2012 Nov.
Article En | MEDLINE | ID: mdl-23076807

Chagas disease caused by Trypanosoma cruzi remains an important neglected tropical disease and a cause of significant morbidity and mortality. No longer confined to endemic areas of Latin America, it is now found in non-endemic areas due to immigration. The parasite may persist in any tissue, but in recent years, there has been increased recognition of adipose tissue both as an early target of infection and a reservoir of chronic infection. The major complications of this disease are cardiomyopathy and megasyndromes involving the gastrointestinal tract. The pathogenesis of Chagas disease is complex and multifactorial involving many interactive pathways. The significance of innate immunity, including the contributions of cytokines, chemokines, reactive oxygen species, and oxidative stress, has been emphasized. The role of the components of the eicosanoid pathway such as thromboxane A(2) and the lipoxins has been demonstrated to have profound effects as both pro- and anti-inflammatory factors. Additionally, we discuss the vasoconstrictive actions of thromboxane A(2) and endothelin-1 in Chagas disease. Human immunity to T. cruzi infection and its role in pathogen control and disease progression have not been fully investigated. However, recently, it was demonstrated that a reduction in the anti-inflammatory cytokine IL-10 was associated with clinically significant chronic chagasic cardiomyopathy.


Chagas Disease/immunology , Trypanosoma cruzi/immunology , Adaptive Immunity , Animals , Chagas Disease/epidemiology , Chagas Disease/pathology , Humans , Immunity, Innate , Life Cycle Stages , Trypanosoma cruzi/growth & development
14.
Am J Pathol ; 181(1): 130-40, 2012 Jul.
Article En | MEDLINE | ID: mdl-22658486

Infection with Trypanosoma cruzi induces inflammation, which limits parasite proliferation but may result in chagasic heart disease. Suppressor of cytokine signaling 2 (SOCS2) is a regulator of immune responses and may therefore participate in the pathogenesis of T. cruzi infection. SOCS2 is expressed during T. cruzi infection, and its expression is partially reduced in infected 5-lipoxygenase-deficient [knockout (KO)] mice. In SOCS2 KO mice, there was a reduction in both parasitemia and the expression of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), IL-6, IL-10, SOCS1, and SOCS3 in the spleen. Expression of IFN-γ, TNF-α, SOCS1, and SOCS3 was also reduced in the hearts of infected SOCS2 KO mice. There was an increase in the generation and expansion of T regulatory (Treg) cells and a decrease in the number of memory cells in T. cruzi-infected SOCS2 KO mice. Levels of lipoxinA(4) (LXA(4)) increased in these mice. Echocardiography studies demonstrated an impairment of cardiac function in T. cruzi-infected SOCS2 KO mice. There were also changes in calcium handling and in action potential waveforms, and reduced outward potassium currents in isolated cardiac myocytes. Our data suggest that reductions of inflammation and parasitemia in infected SOCS2-deficient mice may be secondary to the increases in Treg cells and LXA(4) levels. This occurs at the cost of greater infection-associated heart dysfunction, highlighting the relevance of balanced inflammatory and immune responses in preventing severe T. cruzi-induced disease.


Chagas Cardiomyopathy/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Acute Disease , Animals , Arachidonate 5-Lipoxygenase/physiology , Cells, Cultured , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/pathology , Chagas Cardiomyopathy/physiopathology , Cytokines/biosynthesis , Disease Models, Animal , Heart/parasitology , Lipoxins/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/immunology , Parasite Load , Parasitemia/immunology , Patch-Clamp Techniques , Suppressor of Cytokine Signaling Proteins/deficiency , T-Lymphocyte Subsets/immunology , Trypanosoma cruzi/isolation & purification
15.
Adv Exp Med Biol ; 729: 65-82, 2012.
Article En | MEDLINE | ID: mdl-22411314

The role of caveolin and caveolae in the pathogenesis of infection has only recently been appreciated. In this chapter, we have highlighted some important new data on the role of caveolin in infections due to bacteria, viruses and fungi but with particular emphasis on the protozoan parasites Leishmania spp., Trypanosoma cruzi and Toxoplasma gondii. This is a continuing area of research and the final chapter has not been written on this topic.


Caveolins/metabolism , Host-Pathogen Interactions , Infections/metabolism , Animals , Humans , Infections/microbiology , Infections/parasitology , Infections/virology
16.
Front Biosci (Elite Ed) ; 4(5): 1743-58, 2012 01 01.
Article En | MEDLINE | ID: mdl-22201990

Trypanosoma cruzi is the etiologic agent of Chagas disease. The contributions of parasite and immune system for disease pathogenesis remain unresolved and controversial. The possibility that Chagas disease was an autoimmune progression triggered by T. cruzi infection led some to question the benefit of treating chronically T. cruzi-infected persons with drugs. Furthermore, it provided the rationale for not investing in research aimed at a vaccine which might carry a risk of inducing autoimmunity or exacerbating inflammation. This viewpoint was adopted by cash-strapped health systems in the developing economies where the disease is endemic and has been repeatedly challenged by researchers and clinicians in recent years and there is now a considerable body of evidence and broad consensus that parasite persistence is requisite for pathogenesis and that antiparasitic immunity can be protective against T. cruzi pathogenesis without eliciting autoimmune pathology. Thus, treatment of chronically infected patients is likely to yield positive outcomes and efforts to understand immunity and vaccine development should be recognized as a priority area of research for Chagas disease.


Chagas Disease/pathology , Animals , Antibody Formation , Autoimmunity , Chagas Disease/immunology , Chemokines/physiology , Chronic Disease , Cytokines/physiology , Disease Models, Animal , Humans , Immunity, Cellular , Mice , Toll-Like Receptors/physiology
17.
Adv Parasitol ; 76: 235-50, 2011.
Article En | MEDLINE | ID: mdl-21884894

Adipose tissue is the largest endocrine organ in the body and is composed primarily of adipocytes (fat cells) but also contains fibroblasts, endothelial cells, smooth muscle cells, macrophages and lymphocytes. Adipose tissue and the adipocyte are important in the regulation of energy metabolism and of the immune response. Adipocytes also synthesize adipokines such as adiponectin which is important in the regulation of insulin sensitivity and inflammation. Infection of mice with Trypanosoma cruzi results in an upregulation of inflammation in adipose tissue that begins during the acute phase of infection and persists into the chronic phase. The adipocyte is both a target of infection and a reservoir for the parasite during the chronic phase from which recrudescence of the infection may occur during periods of immunosuppression.


Adipose Tissue/physiopathology , Chagas Disease/immunology , Chagas Disease/pathology , Diabetes Mellitus/chemically induced , Trypanosoma cruzi/immunology , Trypanosoma cruzi/pathogenicity , Adipocytes/physiology , Adipose Tissue/immunology , Animals , Humans
18.
Mem Inst Oswaldo Cruz ; 104 Suppl 1: 219-25, 2009 Jul.
Article En | MEDLINE | ID: mdl-19753477

Trypanosoma cruzi infection of the adipose tissue of mice triggers the local expression of inflammatory mediators and a reduction in the expression of the adipokine adiponectin. T. cruzi can be detected in adipose tissue by PCR 300 days post-infection. Infection of cultured adipocytes results in increased expression of cytokines and chemokines and a reduction in the expression of adiponectin and the peroxisome proliferator-activated receptor gamma, both of which are negative regulators of inflammation. Infection also results in the upregulation of cyclin D1, the Notch pathway, and extracellular signal-regulated kinase and a reduction in the expression of caveolin-1. Thus, T. cruzi infection of cultured adipocytes leads to an upregulation of the inflammatory process. Since adiponectin null mice have a cardiomyopathic phenotype, it is possible that the reduction in adiponectin contributes to the pathogenesis of chagasic cardiomyopathy. Adipose tissue may serve as a reservoir for T. cruzi from which parasites can become reactivated during periods of immunosuppression. T. cruzi infection of mice often results in hypoglycemia. In contrast, hyperglycemia as observed in diabetes results in increased parasitemia and mortality. Adipose tissue is an important target tissue of T. cruzi and the infection of this tissue is associated with a profound impact on systemic metabolism, increasing the risk of metabolic syndrome.


Adipocytes/parasitology , Adipose Tissue/parasitology , Chagas Disease/metabolism , Metabolic Syndrome/parasitology , Adiponectin/metabolism , Adipose Tissue/metabolism , Animals , Disease Models, Animal , Metabolic Syndrome/metabolism , PPAR gamma/metabolism
19.
Mem. Inst. Oswaldo Cruz ; 104(supl.1): 1-7, July 2009. ilus, graf
Article En | LILACS | ID: lil-520882

Trypanosoma cruzi infection of the adipose tissue of mice triggers the local expression of inflammatory mediators and a reduction in the expression of the adipokine adiponectin. T. cruzi can be detected in adipose tissue by PCR 300 days post-infection. Infection of cultured adipocytes results in increased expression of cytokines and chemokines and a reduction in the expression of adiponectin and the peroxisome proliferator-activated receptor ³, both of which are negative regulators of inflammation. Infection also results in the upregulation of cyclin D1, the Notch pathway, and extracellular signal-regulated kinase and a reduction in the expression of caveolin-1. Thus, T. cruzi infection of cultured adipocytes leads to an upregulation of the inflammatory process. Since adiponectin null mice have a cardiomyopathic phenotype, it is possible that the reduction in adiponectin contributes to the pathogenesis of chagasic cardiomyopathy. Adipose tissue may serve as a reservoir for T. cruzi from which parasites can become reactivated during periods of immunosuppression. T. cruzi infection of mice often results in hypoglycemia. In contrast, hyperglycemia as observed in diabetes results in increased parasitemia and mortality. Adipose tissue is an important target tissue of T. cruzi and the infection of this tissue is associated with a profound impact on systemic metabolism, increasing the risk of metabolic syndrome.


Animals , Adipocytes/parasitology , Adipose Tissue/parasitology , Chagas Disease/metabolism , Metabolic Syndrome/parasitology , Adiponectin/metabolism , Adipose Tissue/metabolism , Disease Models, Animal , Metabolic Syndrome/metabolism , PPAR gamma/metabolism
20.
Microbes Infect ; 10(14-15): 1558-66, 2008.
Article En | MEDLINE | ID: mdl-18951994

An intense inflammatory process is associated with Trypanosoma cruzi infection. We investigated the mediators that trigger leukocyte activation and migration to the heart of infected mice. It is known that nitric oxide (NO) modulates the inflammatory response. During T. cruzi infection, increased concentrations of NO are produced by cardiac myocytes (CMs) in response to IFN-gamma and TNF. Here, we investigated whether NO, IFN-gamma and TNF regulate chemokine production by T. cruzi-infected CMs. In addition, we examined the effects of the NOS2 deficiency on chemokine expression both in cultured CMs and in hearts obtained from infected mice. After infection of cultured WT CMs with T. cruzi, the addition of IFN-gamma and TNF increased both mRNA and protein levels of the chemokines CXCL1, CXCL2, CCL2, CCL3, CCL4 and CCL5. Interestingly, T. cruzi-infected NOS2-deficient CMs produced significantly higher levels of CCL2, CCL4, CCL5 and CXL2 in the presence of IFN-gamma and TNF. Infection of NOS2-null mice resulted in a significant increase in the expression of both chemokine mRNA and protein levels in the heart of, compared with hearts obtained from, infected WT mice. Our data indicate that NOS2 is a potent modulator of chemokine expression which is critical to triggering the generation of the inflammatory infiltrate in the heart during T. cruzi infection.


Chemokines/biosynthesis , Myocytes, Cardiac/immunology , Myocytes, Cardiac/parasitology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Trypanosoma cruzi/microbiology , Animals , Cells, Cultured , Gene Expression Profiling , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Myocardium/pathology , Nitric Oxide Synthase Type II/deficiency , Tumor Necrosis Factor-alpha/immunology
...