Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
PLoS Negl Trop Dis ; 6(5): e1644, 2012.
Article in English | MEDLINE | ID: mdl-22590660

ABSTRACT

BACKGROUND: The factors contributing to chronic Chagas' heart disease remain unknown. High nitric oxide (NO) levels have been shown to be associated with cardiomyopathy severity in patients. Further, NO produced via inducible nitric oxide synthase (iNOS/NOS2) is proposed to play a role in Trypanosoma cruzi control. However, the participation of iNOS/NOS2 and NO in T. cruzi control and heart injury has been questioned. Here, using chronically infected rhesus monkeys and iNOS/NOS2-deficient (Nos2(-/-)) mice we explored the participation of iNOS/NOS2-derived NO in heart injury in T. cruzi infection. METHODOLOGY: Rhesus monkeys and C57BL/6 and Nos2(-/-) mice were infected with the Colombian T. cruzi strain. Parasite DNA was detected by polymerase chain reaction, T. cruzi antigens and iNOS/NOS2(+) cells were immunohistochemically detected in heart sections and NO levels in serum were determined by Griess reagent. Heart injury was assessed by electrocardiogram (ECG), echocardiogram (ECHO), creatine kinase heart isoenzyme (CK-MB) activity levels in serum and connexin 43 (Cx43) expression in the cardiac tissue. RESULTS: Chronically infected monkeys presented conduction abnormalities, cardiac inflammation and fibrosis, which resembled the spectrum of human chronic chagasic cardiomyopathy (CCC). Importantly, chronic myocarditis was associated with parasite persistence. Moreover, Cx43 loss and increased CK-MB activity levels were primarily correlated with iNOS/NOS2(+) cells infiltrating the cardiac tissue and NO levels in serum. Studies in Nos2(-/-) mice reinforced that the iNOS/NOS2-NO pathway plays a pivotal role in T. cruzi-elicited cardiomyocyte injury and in conduction abnormalities that were associated with Cx43 loss in the cardiac tissue. CONCLUSION: T. cruzi-infected rhesus monkeys reproduce features of CCC. Moreover, our data support that in T. cruzi infection persistent parasite-triggered iNOS/NOS2 in the cardiac tissue and NO overproduction might contribute to CCC severity, mainly disturbing of the molecular pathway involved in electrical synchrony. These findings open a new avenue for therapeutic tools in Chagas' heart disease.


Subject(s)
Chagas Cardiomyopathy/pathology , Myocardium/enzymology , Nitric Oxide Synthase Type II/analysis , Nitric Oxide/blood , Serum/chemistry , Trypanosoma cruzi/pathogenicity , Animals , Chagas Cardiomyopathy/parasitology , Connexin 43/analysis , Creatine Kinase/blood , Disease Models, Animal , Echocardiography , Electrocardiography , Female , Humans , Immunohistochemistry , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology
2.
PLoS Pathog ; 8(4): e1002645, 2012.
Article in English | MEDLINE | ID: mdl-22532799

ABSTRACT

In Chagas disease, CD8(+) T-cells are critical for the control of Trypanosoma cruzi during acute infection. Conversely, CD8(+) T-cell accumulation in the myocardium during chronic infection may cause tissue injury leading to chronic chagasic cardiomyopathy (CCC). Here we explored the role of CD8(+) T-cells in T. cruzi-elicited heart injury in C57BL/6 mice infected with the Colombian strain. Cardiomyocyte lesion evaluated by creatine kinase-MB isoenzyme activity levels in the serum and electrical abnormalities revealed by electrocardiogram were not associated with the intensity of heart parasitism and myocarditis in the chronic infection. Further, there was no association between heart injury and systemic anti-T. cruzi CD8(+) T-cell capacity to produce interferon-gamma (IFNγ) and to perform specific cytotoxicity. Heart injury, however, paralleled accumulation of anti-T. cruzi cells in the cardiac tissue. In T. cruzi infection, most of the CD8(+) T-cells segregated into IFNγ(+) perforin (Pfn)(neg) or IFNγ(neg)Pfn(+) cell populations. Colonization of the cardiac tissue by anti-T. cruzi CD8(+)Pfn(+) cells paralleled the worsening of CCC. The adoptive cell transfer to T. cruzi-infected cd8(-/-) recipients showed that the CD8(+) cells from infected ifnγ(-/-)pfn(+/+) donors migrate towards the cardiac tissue to a greater extent and caused a more severe cardiomyocyte lesion than CD8(+) cells from ifnγ(+/+)pfn(-/-) donors. Moreover, the reconstitution of naïve cd8(-/-) mice with CD8(+) cells from naïve ifnγ(+/+)pfn(-/-) donors ameliorated T. cruzi-elicited heart injury paralleled IFNγ(+) cells accumulation, whereas reconstitution with CD8(+) cells from naïve ifnγ(-/-)pfn(+/+) donors led to an aggravation of the cardiomyocyte lesion, which was associated with the accumulation of Pfn(+) cells in the cardiac tissue. Our data support a possible antagonist effect of CD8(+)Pfn(+) and CD8(+)IFNγ(+) cells during CCC. CD8(+)IFNγ(+) cells may exert a beneficial role, whereas CD8(+)Pfn(+) may play a detrimental role in T. cruzi-elicited heart injury.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chagas Cardiomyopathy/immunology , Gene Expression Regulation/immunology , Interferon-gamma/immunology , Myocardium/immunology , Pore Forming Cytotoxic Proteins/immunology , Trypanosoma cruzi/immunology , Acute Disease , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Movement/genetics , Cell Movement/immunology , Chagas Cardiomyopathy/genetics , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Female , Gene Expression Regulation/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Male , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pore Forming Cytotoxic Proteins/biosynthesis , Pore Forming Cytotoxic Proteins/genetics , Trypanosoma cruzi/metabolism
3.
Int J Exp Pathol ; 91(1): 72-86, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19878357

ABSTRACT

Understanding the dual participation of the immune response in controlling the invader and at the same time causing tissue damage might contribute to the design of effective new vaccines and therapies for Chagas disease. Perforin, a cytolytic protein product of killer cells, is involved in resistance to acute Trypanosoma cruzi infection. However, the contribution of perforin in parasite control and chronic chagasic cardiomyopathy is unclear. Perforin-positive cells were detected in the heart tissue during the acute and chronic phases of infection of C57BL/6 mice inoculated with low dose (10(2) parasites) of the Colombian T. cruzi strain. This protocol led to acute phase survival in both wild-type and perforin null (pfp(-/-)) mice lineages. During the chronic infection, parasitism and inducible nitric oxide synthase (iNOS) as well as interleukin (IL)-4+ and, mainly, interferon (IFN)-gamma+ cells were more elevated in the heart tissue of pfp(-/-) mice. Higher levels of circulating NO and anti-parasite immunoglobulin (Ig)G2c and IgG3, paralleled by a prominent frequency of IFN-gamma+ and IL-10+ splenocytes, were present in pfp(-/-)-infected mice. Therefore, although the perforin-dependent pathway plays a role, it is not crucial for anti-T. cruzi immunity and acute phase survival of mice infected with a low inoculum. Further, perforin deficiency resulted in lower activity of creatine kinase-muscle brain isoform (CK-MB) isoenzyme in serum and a more restricted connexin 43 loss, both of which are markers of the cardiomyocyte lesion. Moreover, perforin deficiency hampered the development of severe electrocardiographic abnormalities. Hence, our results corroborate that perforin-bearing cytotoxic cells might contribute to cardiomyocyte lesion and heart dysfunction during chronic T. cruzi infection, shedding light on immunopathogenesis of chronic chagasic cardiomyopathy.


Subject(s)
Chagas Cardiomyopathy/immunology , Cytotoxicity, Immunologic , Myocarditis/immunology , Myocardium/immunology , Pore Forming Cytotoxic Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Trypanosoma cruzi/immunology , Acute Disease , Animals , Antibodies, Protozoan/blood , Cells, Cultured , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/physiopathology , Chagas Cardiomyopathy/prevention & control , Chronic Disease , Connexin 43/metabolism , Creatine Kinase, MB Form/blood , Disease Models, Animal , Electrocardiography , Female , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/parasitology , Myocarditis/physiopathology , Myocarditis/prevention & control , Myocardium/metabolism , Nitric Oxide/blood , Nitric Oxide Synthase Type II/metabolism , Pore Forming Cytotoxic Proteins/deficiency , Pore Forming Cytotoxic Proteins/genetics , T-Lymphocytes, Cytotoxic/parasitology , Time Factors , Trypanosoma cruzi/pathogenicity
4.
Mem Inst Oswaldo Cruz ; 104 Suppl 1: 226-35, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19753478

ABSTRACT

One hundred years ago, Carlos Chagas discovered a new disease, the American trypanosomiasis. Chagas and co-workers later characterised the disease's common manifestation, chronic cardiomyopathy, and suggested that parasitic persistence coupled with inflammation was the key underlying pathogenic mechanism. Better comprehension of the molecular mechanisms leading to clinical heart afflictions is a prerequisite to developing new therapies that ameliorate inflammation and improve heart function without hampering parasite control. Here, we review recent data showing that distinct cell adhesion molecules, chemokines and chemokine receptors participate in anti-parasite immunity and/or detrimental leukocyte trafficking to the heart. Moreover, we offer evidence that CC-chemokine receptors may be attractive therapeutic targets aiming to regain homeostatic balance in parasite/host interaction thereby improving prognosis, supporting that it is becoming a non-phantasious proposal.


Subject(s)
Cell Adhesion Molecules/immunology , Chagas Cardiomyopathy/immunology , Myocarditis/immunology , Receptors, Chemokine/immunology , Trypanosoma cruzi/immunology , Animals , CD4-CD8 Ratio , Cell Movement , Chagas Cardiomyopathy/therapy , Chronic Disease , Myocarditis/parasitology , Trypanosoma cruzi/pathogenicity
5.
Mem. Inst. Oswaldo Cruz ; 104(supl.1): 226-235, July 2009. ilus, graf
Article in English | LILACS | ID: lil-520883

ABSTRACT

One hundred years ago, Carlos Chagas discovered a new disease, the American trypanosomiasis. Chagas and co-workers later characterised the disease's common manifestation, chronic cardiomyopathy, and suggested that parasitic persistence coupled with inflammation was the key underlying pathogenic mechanism. Better comprehension of the molecular mechanisms leading to clinical heart afflictions is a prerequisite to developing new therapies that ameliorate inflammation and improve heart function without hampering parasite control. Here, we review recent data showing that distinct cell adhesion molecules, chemokines and chemokine receptors participate in anti-parasite immunity and/or detrimental leukocyte trafficking to the heart. Moreover, we offer evidence that CC-chemokine receptors may be attractive therapeutic targets aiming to regain homeostatic balance in parasite/host interaction thereby improving prognosis, supporting that it is becoming a non-phantasious proposal.


Subject(s)
Animals , Cell Adhesion Molecules/immunology , Chagas Cardiomyopathy/immunology , Myocarditis/immunology , Receptors, Chemokine/immunology , Trypanosoma cruzi/immunology , Cell Movement , Chronic Disease , Chagas Cardiomyopathy/therapy , Myocarditis/parasitology , Trypanosoma cruzi/pathogenicity
6.
Mem Inst Oswaldo Cruz ; 103(4): 375-85, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18660993

ABSTRACT

In Chagas disease, understanding how the immune response controls parasite growth but also leads to heart damage may provide insight into the design of new therapeutic strategies. Tumor necrosis factor-alpha (TNF-alpha) is important for resistance to acute Trypanosoma cruzi infection; however, in patients suffering from chronic T. cruzi infection, plasma TNF-alpha levels correlate with cardiomyopathy. Recent data suggest that CD8-enriched chagasic myocarditis formation involves CCR1/CCR5-mediated cell migration. Herein, the contribution of TNF-alpha, especially signaling through the receptor TNFR1/p55, to the pathophysiology of T. cruzi infection was evaluated with a focus on the development of myocarditis and heart dysfunction. Colombian strain-infected C57BL/6 mice had increased frequencies of TNFR1/p55+ and TNF-alpha+ splenocytes. Although TNFR1-/- mice exhibited reduced myocarditis in the absence of parasite burden, they succumbed to acute infection. Similar to C57BL/6 mice, Benznidazole-treated TNFR1-/- mice survived acute infection. In TNFR1-/- mice, reduced CD8-enriched myocarditis was associated with defective activation of CD44+CD62Llow/- and CCR5+ CD8+ lymphocytes. Also, anti-TNF-alpha treatment reduced the frequency of CD8+CCR5+ circulating cells and myocarditis, though parasite load was unaltered in infected C3H/HeJ mice. TNFR1-/- and anti-TNF-alpha-treated infected mice showed regular expression of connexin-43 and reduced fibronectin deposition, respectively. Furthermore, anti-TNF-alpha treatment resulted in lower levels of CK-MB, a cardiomyocyte lesion marker. Our results suggest that TNF/TNFR1 signaling promotes CD8-enriched myocarditis formation and heart tissue damage, implicating the TNF/TNFR1 signaling pathway as a potential therapeutic target for control of T. cruzi-elicited cardiomyopathy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/immunology , Chagas Cardiomyopathy/immunology , Receptors, CCR5/immunology , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Cell Movement , Chagas Cardiomyopathy/drug therapy , Chronic Disease , Female , Flow Cytometry , Immunohistochemistry , Infliximab , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Tumor Necrosis Factor, Type I/blood , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
7.
Mem. Inst. Oswaldo Cruz ; 103(4): 375-385, June 2008. ilus, graf, tab
Article in English | LILACS | ID: lil-486867

ABSTRACT

In Chagas disease, understanding how the immune response controls parasite growth but also leads to heart damage may provide insight into the design of new therapeutic strategies. Tumor necrosis factor-alpha (TNF-á) is important for resistance to acute Trypanosoma cruzi infection; however, in patients suffering from chronic T. cruzi infection, plasma TNF-á levels correlate with cardiomyopathy. Recent data suggest that CD8-enriched chagasic myocarditis formation involves CCR1/CCR5-mediated cell migration. Herein, the contribution of TNF-á, especially signaling through the receptor TNFR1/p55, to the pathophysiology of T. cruzi infection was evaluated with a focus on the development of myocarditis and heart dysfunction. Colombian strain-infected C57BL/6 mice had increased frequencies of TNFR1/p55+ and TNF-á+ splenocytes. Although TNFR1-/- mice exhibited reduced myocarditis in the absence of parasite burden, they succumbed to acute infection. Similar to C57BL/6 mice, Benznidazole-treated TNFR1-/- mice survived acute infection. In TNFR1-/- mice, reduced CD8-enriched myocarditis was associated with defective activation of CD44+CD62Llow/- and CCR5+ CD8+ lymphocytes. Also, anti-TNF-á treatment reduced the frequency of CD8+CCR5+ circulating cells and myocarditis, though parasite load was unaltered in infected C3H/HeJ mice. TNFR1-/- and anti-TNF-á-treated infected mice showed regular expression of connexin-43 and reduced fibronectin deposition, respectively. Furthermore, anti-TNF-á treatment resulted in lower levels of CK-MB, a cardiomyocyte lesion marker. Our results suggest that TNF/TNFR1 signaling promotes CD8-enriched myocarditis formation and heart tissue damage, implicating the TNF/TNFR1 signaling pathway as a potential therapeutic target for control of T. cruzi-elicited cardiomyopathy.


Subject(s)
Animals , Female , Mice , Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , /immunology , Chagas Cardiomyopathy/immunology , /immunology , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Cell Movement , Chronic Disease , Chagas Cardiomyopathy/drug therapy , Flow Cytometry , Immunohistochemistry , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Tumor Necrosis Factor, Type I/blood , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
8.
Rio de Janeiro; s.n; 2007. xi,107 p. tab, graf, ilus.
Thesis in Portuguese | LILACS | ID: lil-481747

ABSTRACT

Dentre os mecanismos efetores utilizados pelo sistema imunológico contra o T. cruzi, podemos destacar os linfócitos T CD8 como células críticas na resistência ao parasito e também na patogênese da miocardite e da cardiopatia crônica. Os linfócitos T CD8 medeiam esta proteção pela secreção de citocinas, principalmente de perfil Th1, e pela atividade citotóxica através de moléculas citolíticas tais como perforina. Na infecção pelo T. cruzi tem-se observado que a perforina participa do controle do parasito e/ou da imunopatogênese da infecção por mecanismos não completamente entendidos. Inicialmente, demonstramos o aumento da expressão da perforina no tecido cardíaco de animais C57BL/6 aguda e cronicamente infectados pelo T. cruzi, o que ocasionou nosso interesse em estudar o envolvimento da perforina no controle do parasito, na formação do infiltrado inflamatório e geração da lesão cardíaca e conseqüentemente cardiopatia. Nas etapas iniciais da infecção, animais deficientes em perforina (PKO) infectados com 100 parasitos apresentaram aumento da parasitemia associado ao aumento sérico significativo de óxido nítrico e dos anticorpos IgG2a e IgG3. Além disso, houve um controle da parasitemia na fase crônica, de forma similar aos animais C57BL/6. A análise ex vivo de citocinas Th1 (IFNy) e Th2 (IL-4) revelou um aumento significativo de produção de IFNy pelos linfócitos isolados do baço de animais PKO infectados, quando comparado aos animais C57BL/6. Além disso, animais PKO controle apresentaram níveis basais aumentados de IL-4. É possível que o aumento basal de células produtoras de IL-4 nos animais PKO estejam modulando o controle do parasito e a resistência à infecção. De forma interessante, a constituição do infiltrado inflamatório foi distinta entre os animais C57BL/6 e PKO. No tecido cardíaco dos animais PKO observamos na fase aguda de infecção diminuição do número de células CD8(mais), aumento do número de células iNOS(mais) e de células produtoras...cardíacas.


Subject(s)
Animals , Chagas Disease , Parasitic Diseases, Animal , Trypanosoma cruzi
SELECTION OF CITATIONS
SEARCH DETAIL
...