Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Immun Inflamm Dis ; 5(2): 163-176, 2017 06.
Article in English | MEDLINE | ID: mdl-28474508

ABSTRACT

INTRODUCTION: Complete Freund's Adjuvant (CFA) emulsified with an antigen is a widely used method to induce autoimmune disease in animal models, yet the contribution of CFA to the immune response is not well understood. We compared the effectiveness of CFA with Incomplete Freund's Adjuvant (IFA) or TiterMax Gold Adjuvant (TMax) in experimental autoimmune myocarditis (EAM) in male mice. METHODS: EAM was induced in A/J, BALB/c, and IL6KO BALB/c male mice by injection of the myocarditogenic peptide in CFA, IFA, or TMax on days 0 and 7. EAM severity was analyzed by histology on day 21. In addition, specific flow cytometry outcomes were evaluated on day 21. RESULTS: Only mice immunized with CFA and myocarditogenic peptide on both days 0 and 7 developed substantial myocarditis as measured by histology. We observed a significantly increased level of IL6 in the spleen 3 days after CFA immunization. In the spleen and heart on day 21, there was an expansion of myeloid cells in CFA-immunized mice, as compared to IFA or TMax-immunized animals. Recombinant IL-6 at the time of IFA immunization partially restored susceptibility of the mice to EAM. We also treated EAM-resistant IL-6 knockout mice with recombinant IL-6 around the time of the first immunization, on days -1 to 2, completely restoring disease susceptibility, showing that the requirement for IL-6 coincides with primary immunization. Examining APC populations in the lymph node draining the immunization site evidenced the contribution of IL-6 to the CFA-dependence of EAM was through controlling local dendritic cell (DC) trafficking. CONCLUSIONS: CFA used with myocarditogenic peptide twice is required to induce EAM in both A/J and Balb/c mice. Although IFA and TiterMax induce antibody responses, only CFA preferentially induced autoantigen-specific responses. CFA expands monocytes in the heart and in the spleen. IL-6 signaling is required during short window around primary immunization to induce EAM. In addition, IL-6 deficient mice resistance to EAM could be reversed by injecting IL-6 around first immunization. IL-6 expands dendritic cell and monocytic populations and ultimately leads to a robust T-cell driven immune response in CFA immunized mice.


Subject(s)
Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Freund's Adjuvant/adverse effects , Interleukin-6/immunology , Myocarditis/chemically induced , Myocarditis/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Freund's Adjuvant/pharmacology , Interleukin-6/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Myocarditis/genetics , Myocarditis/pathology
2.
Cytokine ; 74(1): 62-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25649043

ABSTRACT

IL6 is a pleiotropic cytokine that is made in response to perturbations in homeostasis. IL6 becomes elevated in the acute response to host injury and can activate immune cells, direct immune cell trafficking, signal protective responses in local tissue, initial the acute phase response or initiate wound healing. In the short term this proinflammatory response is protective and limits host damage. It is when this acute response remains chronically activated that IL6 becomes pathogenic to the host. Chronically elevated IL6 levels lead to chronic inflammation and fibrotic disorders. The heart is a tissue where this temporal regulation of IL6 is very apparent. Studies from myocardial infarction show how short-term IL6 signaling can protect and preserve the heart tissue in response to acute damage, where long term IL6 signaling or an over-production of IL6R protein plays a causal role in cardiovascular disease. Thus, IL6 can be both protective and pathogenic, depending on the kinetics of the host response.


Subject(s)
Heart Diseases/immunology , Heart Diseases/metabolism , Heart Failure/immunology , Heart Failure/metabolism , Interleukin-6/metabolism , Myocardium/metabolism , Animals , Cytokine Receptor gp130/immunology , Cytokine Receptor gp130/metabolism , Humans , Inflammation/immunology , Interleukin-6/biosynthesis , Myocardial Infarction/etiology , Myocardial Infarction/immunology , Myocarditis/immunology , Myocardium/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction
3.
Am J Pathol ; 185(3): 847-61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25622543

ABSTRACT

Myocarditis is a leading cause of sudden cardiac failure in young adults. Natural killer (NK) cells, a subset of the innate lymphoid cell compartment, are protective in viral myocarditis. Herein, we demonstrated that these protective qualities extend to suppressing autoimmune inflammation. Experimental autoimmune myocarditis (EAM) was initiated in BALB/c mice by immunization with myocarditogenic peptide. During EAM, activated cardiac NK cells secreted interferon γ, perforin, and granzyme B, and expressed CD69, tumor necrosis factor-related apoptosis-inducing ligand treatment, and CD27 on their cell surfaces. The depletion of NK cells during EAM with anti-asialo GM1 antibody significantly increased myocarditis severity, and was accompanied by elevated fibrosis and a 10-fold increase in the percentage of cardiac-infiltrating eosinophils. The resultant influx of eosinophils to the heart was directly responsible for the increased disease severity in the absence of NK cells, because treatment with polyclonal antibody asialogangloside GM-1 did not augment myocarditis severity in eosinophil-deficient ΔdoubleGATA1 mice. We demonstrate that NK cells limit eosinophilic infiltration both indirectly, through altering eosinophil-related chemokine production by cardiac fibroblasts, and directly, by inducing eosinophil apoptosis in vitro. Altogether, we define a new pathway of eosinophilic regulation through interactions with NK cells.


Subject(s)
Eosinophils/immunology , Killer Cells, Natural/immunology , Myocarditis/immunology , Myocardium/immunology , Animals , Apoptosis/immunology , Eosinophils/pathology , Fibrosis/immunology , Fibrosis/pathology , Inflammation/pathology , Killer Cells, Natural/pathology , Mice , Mice, Inbred BALB C , Myocarditis/pathology , Myocardium/pathology
4.
J Immunol ; 191(8): 4038-47, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048893

ABSTRACT

CD4(+) T cells play a central role in inflammatory heart disease, implicating a cytokine product associated with Th cell effector function as a necessary mediator of this pathophysiology. IFN-γ-deficient mice developed severe experimental autoimmune myocarditis (EAM), in which mice are immunized with cardiac myosin peptide, whereas IL-17A-deficient mice were protected from progression to dilated cardiomyopathy. We generated IFN-γ(-/-)IL-17A(-/-) mice to assess whether IL-17 signaling was responsible for the severe EAM of IFN-γ(-/-) mice. Surprisingly, IFN-γ(-/-)IL-17A(-/-) mice developed a rapidly fatal EAM. Eosinophils constituted a third of infiltrating leukocytes, qualifying this disease as eosinophilic myocarditis. We found increased cardiac production of CCL11/eotaxin, as well as Th2 deviation, among heart-infiltrating CD4(+) cells. Ablation of eosinophil development improved survival of IFN-γ(-/-)IL-17A(-/-) mice, demonstrating the necessity of eosinophils in fatal heart failure. The severe and rapidly fatal autoimmune inflammation that developed in the combined absence of IFN-γ and IL-17A constitutes a novel model of eosinophilic heart disease in humans. This is also, to our knowledge, the first demonstration that eosinophils have the capacity to act as necessary mediators of morbidity in an autoimmune process.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Eosinophils/immunology , Interferon-gamma/deficiency , Interleukin-17/deficiency , Myocarditis/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Biomarkers , Cardiac Myosins/immunology , Cardiomyopathies/immunology , Chemokine CCL11/biosynthesis , Inflammation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Myocarditis/genetics , Myocarditis/prevention & control , Myocardium/immunology , Myositis
SELECTION OF CITATIONS
SEARCH DETAIL
...