Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Basic Res Cardiol ; 107(5): 287, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22836587

ABSTRACT

Enteroviruses, such as coxsackieviruses of group B (CVB), are able to induce a chronic inflammation of the myocardium, which may finally lead to the loss of functional tissue, remodeling processes and the development of fibrosis, thus affecting the proper contractile function of the heart. In other fibrotic diseases like scleroderma, the prostacyclin agonist iloprost was found to inhibit the extracellular signal-regulated kinase (ERK, p44/42 MAPK), a mitogen-activated protein kinase, and consecutively, the expression of the profibrotic cytokine connective tissue growth factor (CTGF), thereby preventing the development of fibrosis. As CTGF was found to mediate fibrosis in chronic CVB3 myocarditis as well, we evaluated whether the in vivo application of iloprost is capable to reduce the development of ERK/CTGF-mediated fibrosis in enteroviral myocarditis. Unexpectedly, the application of iloprost resulted in a prolonged myocardial inflammation and an aggravated fibrosis and failed to reduce activation of ERK and expression of CTGF at later stages of the disease. In addition, viral replication was found to be increased in iloprost-treated mice. Notably, the expression of cardiac inducible nitric oxide synthase (iNOS), which is known to aggravate myocardial damage in CVB3-infected mice, was strongly enhanced by iloprost. Using cultivated bone marrow macrophages (BMM), we confirmed these results, proving that iloprost potentiates the expression of iNOS mRNA and protein in CVB3-infected and IFN-gamma stimulated BMM. In conclusion, these results suggest a critical reflection of the clinical use of iloprost, especially in patients possibly suffering from an enteroviral myocarditis.


Subject(s)
Enterovirus B, Human , Enterovirus Infections/virology , Extracellular Signal-Regulated MAP Kinases/physiology , Iloprost/pharmacology , MAP Kinase Signaling System/drug effects , Myocarditis/virology , Myocardium/pathology , Nitric Oxide Synthase Type II/genetics , Virus Replication/drug effects , Animals , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Connective Tissue Growth Factor/genetics , Enterovirus Infections/metabolism , Enterovirus Infections/pathology , Fibrosis , MAP Kinase Signaling System/physiology , Mice , Myocarditis/metabolism , Myocarditis/pathology
3.
Amino Acids ; 39(1): 243-55, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19997756

ABSTRACT

Proteasomes are known to be the main suppliers of MHC class I (MHC-I) ligands. In an attempt to identify coxsackievirus B3 (CVB3)-MHC-I epitopes, a combined approach of in silico MHC-I/transporters associated with antigen processing (TAP)-binding and proteasomal cleavage prediction was applied. Accordingly, 13 potential epitopes originating from the structural and non-structural protein region of CVB3 were selected for further in vitro processing analysis by proteasomes. Mass spectrometry demonstrated the generation of seven of the 13 predicted MHC-I ligands or respective ligand precursors by proteasomes. Detailed processing analysis of three adjacent MHC-I ligands with partially overlapping sequences, i.e. VP2(273-281), VP2(284-292) and VP2(285-293), revealed the preferential generation predominantly of the VP2(285-293) epitope by immunoproteasomes due to altered cleavage site preferences. The VP2(285-293) peptide was identified to be a high affinity binder, rendering VP2(285-293) a likely candidate for CD8 T cell immunity in CVB3 infection. In conclusion, the concerted usage of different in silico prediction methods and in vitro epitope processing/presentation studies was supportive in the identification of CVB3 MHC-I epitopes.


Subject(s)
Computational Biology , Enterovirus B, Human/chemistry , Enterovirus B, Human/immunology , Epitopes/immunology , Epitopes/metabolism , Histocompatibility Antigens Class I/immunology , Proteasome Endopeptidase Complex/metabolism , Animals , Ligands , Mice , Mice, Inbred C57BL
4.
Am J Pathol ; 175(2): 510-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19590042

ABSTRACT

Murine models of coxsackievirus B3 (CVB3)-induced myocarditis mimic the divergent human disease course of cardiotropic viral infection, with host-specific outcomes ranging from complete recovery in resistant mice to chronic disease in susceptible hosts. To identify susceptibility factors that modulate the course of viral myocarditis, we show that type-I interferon (IFN) responses are considerably impaired in acute CVB3-induced myocarditis in susceptible mice, which have been linked to immunoproteasome (IP) formation. Here we report that in concurrence with distinctive type-I IFN kinetics, myocardial IP formation peaked early after infection in resistant mice and was postponed with maximum IP expression concomitant to massive inflammation and predominant type-II IFN responses in susceptible mice. IP activity is linked to a strong enhancement of antigenic viral peptide presentation. To investigate the impact of myocardial IPs in CVB3-induced myocarditis, we identified two novel CVB3 T cell epitopes, virus capsid protein 2 [285-293] and polymerase 3D [2170-2177]. Analysis of myocardial IPs in CVB3-induced myocarditis revealed that myocardial IP expression resulted in efficient epitope generation. As opposed to the susceptible host, myocardial IP expression at early stages of disease corresponded to enhanced CVB3 epitope generation in the hearts of resistant mice. We propose that this process may precondition the infected heart for adaptive immune responses. In conclusion, type-I IFN-induced myocardial IP activity at early stages coincides with less severe disease manifestation in CVB3-induced myocarditis.


Subject(s)
Enterovirus Infections/immunology , Enterovirus/immunology , Interferon Type I/immunology , Myocarditis/immunology , Myocarditis/virology , Proteasome Endopeptidase Complex/immunology , Animals , Disease Models, Animal , Enterovirus Infections/complications , Enterovirus Infections/pathology , Epitopes, T-Lymphocyte/immunology , Humans , Interferon Type I/pharmacology , Mice , Mice, Inbred C57BL , Myocarditis/pathology , Proteasome Endopeptidase Complex/drug effects
5.
Circ Res ; 104(7): 851-9, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19246678

ABSTRACT

The characteristics of dilated cardiomyopathy (DCM) resulting from chronic viral myocarditis are remodeling processes of the extracellular matrix. Based on our findings of enhanced osteopontin (OPN) expression in inflamed human hearts, we further investigated in the murine model of acute and chronic coxsackievirus (CV)B3-myocarditis the role of OPN regarding its involvement in resolution of cardiac virus infection and fibrosis. In hearts of A.BY/SnJ mice susceptible to chronic CVB3-myocarditis, a pronounced increase of OPN expression levels was detected by microarray analysis and quantitative RT-PCR during acute stages of myocarditis. Combined immunohistochemistry and in situ hybridization identified infiltrating macrophages as main OPN producers. In contrast to resistant C57BL/6 and OPN gene-deficient mice, transcription levels of matrix metalloproteinase-3, TIMP1 (tissue inhibitor of metalloproteinases-1), uPA (urokinase-type plasminogen activator), and transforming growth factor beta1 were elevated in susceptible mice, and as a consequence, procollagen-1alpha mRNA expression and fibrosis was considerably enhanced. Treatment of infected susceptible mice with the vitamin D analog ZK 191784 led to decreased myocardial expression levels of OPN, metalloproteinase-3, TIMP1, uPA, and procollagen-1alpha and subsequently to reduced fibrosis. Concurrently, the fibrosis-relevant signaling molecules pERK (phosphorylated extracellular signal-regulated kinase) and pAkt (phosphorylated Akt), increased in A.BY/SnJ mice, were diminished in ZK 191784-treated mice. Here, we show that high expression levels of OPN in acute myocarditis are associated with consecutive development of extensive fibrosis that can be reduced by treatment with a vitamin D analog. Thus, OPN may serve as a diagnostic tool as well as a potential therapeutic target to limit cardiac remodeling in chronic myocarditis.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Coxsackievirus Infections/metabolism , Myocarditis/metabolism , Myocardium/metabolism , Osteopontin/metabolism , Ventricular Remodeling , Acute Disease , Animals , Biomarkers/metabolism , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/virology , Chronic Disease , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Coxsackievirus Infections/pathology , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/virology , Disease Models, Animal , Enterovirus B, Human/pathogenicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Humans , Macrophages/metabolism , Macrophages/virology , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/pathology , Myocarditis/physiopathology , Myocarditis/virology , Myocardium/pathology , Osteopontin/deficiency , Osteopontin/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Ventricular Remodeling/drug effects
6.
J Virol ; 82(16): 8149-60, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550677

ABSTRACT

Enteroviruses such as coxsackievirus B3 (CVB3) are able to induce lethal acute and chronic myocarditis. In resistant C57BL/6 mice, CVB3 myocarditis is abrogated by T-cell-dependent mechanisms, whereas major histocompatibility complex (MHC)-matched permissive A.BY/SnJ mice develop chronic myocarditis based on virus persistence. To define the role of T-cell-priming dendritic cells (DCs) in the outcome of CVB3 myocarditis, DCs were analyzed in this animal model in the course of CVB3 infection. In both mouse strains, DCs were found to be infectible with CVB3; however, formation of infectious virions was impaired. In DCs derived from C57BL/6 mice, significantly higher quantities of interleukin-10 (IL-10) and the proinflammatory cytokines IL-6 and tumor necrosis factor alpha were measured compared to those from A.BY/SnJ mice. Additionally, the chemokines interferon-inducible protein 10 (IP-10) and RANTES were secreted by DCs from resistant C57BL/6 mice earlier in infection and at significantly higher levels. The protective role of IP-10 in CVB3 myocarditis was confirmed in IP-10(-/-) mice, which had increased myocardial injury compared to the immunocompetent control animals. Also, major differences in resistant and permissive mice were found in DC subsets, with C57BL/6 mice harboring more cross-priming CD4(-) CD8(+) DCs. As CD4(-) CD8(+) DCs are known to express 10 times more Toll-like receptor 3 (TLR3) than other DC subsets, we followed the course of CVB3 infection in TLR3(-/-) mice. These mice developed a fulminant acute myocarditis and secreted sustained low amounts of type I interferons; secretion of IP-10 and RANTES was nearly abrogated in DCs. We conclude that MHC-independent genetic factors involving DC-related IP-10 secretion and TLR3 expression are beneficial in the prevention of chronic coxsackievirus myocarditis.


Subject(s)
CD4 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , Chemokines/metabolism , Coxsackievirus Infections/virology , Dendritic Cells/metabolism , Myocarditis/virology , Animals , Chemokine CCL5/biosynthesis , Coxsackievirus Infections/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/pathology , Phenotype , Receptors, Cytokine/biosynthesis , Toll-Like Receptor 3/metabolism
7.
J Mol Med (Berl) ; 86(1): 49-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17846733

ABSTRACT

Dilated cardiomyopathy (DCM) as a consequence of viral myocarditis is a worldwide cause of morbidity and death. The deposition of matrix proteins, such as collagen, in the course of ongoing viral myocarditis results in cardiac remodeling and finally in cardiac fibrosis, the hallmark of DCM. To identify mediators of virus-induced cardiac fibrosis, microarray analysis was conducted in a murine model of chronic coxsackievirus B3 (CVB3) myocarditis. By this attempt, we identified connective tissue growth factor (CTGF) as a novel factor highly expressed in infected hearts. Further investigations by quantitative reverse transcription polymerase chain reaction and Western blot analysis confirmed a strong induction of cardiac CTGF expression in the course of CVB3 myocarditis. By in situ hybridization and immunohistochemistry, basal CTGF messenger ribonucleic acid (mRNA) and protein expression were confined in noninfected control hearts mainly to endothelial cells, whereas in CVB3-infected hearts, also numerous fibroblasts were found to express CTGF. Regulation of CTGF is known to be basically mediated by transforming growth factor (TGF)-beta. In the course of CVB3 myocarditis, CTGF upregulation coincided with increased cardiac TGF-beta and procollagen type I mRNA expression, preceding the formation of fibrotic lesions. In in vitro experiments, we found that downregulation of CVB3 replication by means of small interfering RNAs (siRNAs) reverses the upregulation of CTGF mRNA expression. In contrast, downregulation of CTGF by siRNA molecules did not significantly reduce viral load, indicating that CTGF is not essential for CVB3 life cycle. The significantly enhanced transcript levels of TGF-beta, CTGF, and procollagen type I in cultivated CVB3-infected primary cardiac fibroblasts substantiate the role of fibroblasts as a relevant cell population in cardiac remodeling processes. We conclude that CTGF is a crucial molecule in the development of fibrosis in ongoing enteroviral myocarditis. Thus, downregulation of cardiac CTGF expression may open novel therapeutic approaches counteracting the development of cardiac fibrosis and subsequent heart muscle dysfunction.


Subject(s)
Collagen Type I/genetics , Connective Tissue Growth Factor/genetics , Enterovirus Infections/pathology , Myocarditis/pathology , Transforming Growth Factor beta/genetics , Animals , Connective Tissue Growth Factor/analysis , Cytokines , Disease Models, Animal , Endothelial Cells , Fibroblasts/immunology , Fibroblasts/pathology , Mice , Myocarditis/microbiology , Myocardium/chemistry , Myocardium/pathology , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology
8.
Am J Pathol ; 169(6): 2085-93, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17148671

ABSTRACT

Ongoing coxsackievirus B3 (CVB3) myocarditis is characterized by persistence of viral RNA and chronic inflammation primarily mediated by macrophages and T cells. Activated macrophages produce anti-viral effector molecules comprising reactive nitrogen intermediates; however, reactive nitrogen intermediates also contribute to host tissue damage. Controlled activation of macrophages depends on interferon (IFN)-gamma and interleukin (IL)-10. To evaluate mechanisms involved in CVB3-induced pathogenesis of myocarditis, we determined the relationship of inducible nitric-oxide synthase (iNOS) mRNA expression with IFN-gamma and IL-10 secretion during CVB3 infection in different mouse strains. We found in susceptible A.BY/SnJ mice that develop ongoing myocarditis, a low and delayed IFN-gamma secretion and highly diminished IL-10 production compared with resistant C57BL/6 mice. Consequently, iNOS mRNA synthesis was delayed but clearly prolonged in susceptible mice. IL-10 gene-deficient mice confirmed the regulatory role of IL-10 in the outcome of CVB3 myocarditis. These mice did not establish a persistent cardiac infection and revealed IFN-gamma secretion kinetics similar to resistant mice but showed a slightly elongated cardiac iNOS mRNA expression resulting in extended myocarditis. We conclude that coordinated secretion of IFN-gamma and IL-10 is crucial for the effective resolution of CVB3 myocarditis. Moreover, lack of regulatory IL-10 leads to uncontrolled iNOS mRNA production, thus contributing to ongoing myocardial injury.


Subject(s)
Coxsackievirus Infections/pathology , Interleukin-10/metabolism , Myocarditis/pathology , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Animals , Antibody Formation , Coxsackievirus Infections/complications , Coxsackievirus Infections/metabolism , Interferon-gamma/metabolism , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/etiology , Myocarditis/metabolism , Nitric Oxide Synthase , Nitric Oxide Synthase Type II/biosynthesis
9.
Am J Pathol ; 168(5): 1542-52, 2006 May.
Article in English | MEDLINE | ID: mdl-16651621

ABSTRACT

A growing body of evidence indicates that viral infections of the heart contribute to ongoing myocarditis and dilated cardiomyopathy. Murine models of coxsackievirus B3 (CVB3)-induced myocarditis mimic the human disease and allow identification of susceptibility factors that modulate the course of viral myocarditis. Susceptible mouse strains develop chronic myocarditis on the basis of restricted viral replication, whereas resistant strains recover after successful virus elimination. In comparative whole-genome microarray analyses of infected hearts, several genes involved in the processing and presentation of viral epitopes were found to be uniformly up-regulated in acutely CVB3-infected susceptible mice compared with resistant animals. In particular, expression of the catalytic subunits LMP2, LMP7, and MECL-1, immunoproteasome proteins important in the generation of major histocom-patibility complex (MHC) class I-restricted peptides, was clearly enhanced in the susceptible host. Increased expression resulted in enhanced formation of immunoproteasomes and altered proteolytic activities of proteasomes in the heart. This was accompanied by a concerted up-regulation of the antigen-presenting machinery in susceptible mice. Thus, we propose that increased formation of immunoproteasomes in susceptible mice affects the generation of antigenic peptides and the subsequent T-cell-mediated immune responses.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Myocarditis/immunology , Myocardium/metabolism , Proteasome Endopeptidase Complex/physiology , Animals , Antigen Presentation/physiology , Coxsackievirus Infections/metabolism , Disease Models, Animal , Disease Susceptibility , Electrophoresis, Gel, Two-Dimensional , Heart/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Multienzyme Complexes/metabolism , Myocarditis/chemically induced , Myocarditis/etiology , Myocarditis/metabolism , Myocytes, Cardiac/metabolism , Organ Specificity , Peptide Hydrolases/metabolism
10.
Med Microbiol Immunol ; 193(2-3): 101-7, 2004 May.
Article in English | MEDLINE | ID: mdl-12920583

ABSTRACT

Endomyocardial biopsy (EMB) is often performed in patients presenting with sudden onset of heart failure to identify myocarditis. The introduction of immunohistochemical techniques for the detection and differentiation of infiltrating immune cells, specific adhesion molecules and MHC class I and II molecules increased the prognostic value of EMB in the diagnosis of myocarditis considerably. A major breakthrough in the understanding of pathogenetic mechanisms in myocarditis was achieved by diagnostic use of molecular biological methods. By application of in situ hybridization and PCR, enteroviruses, and more recently, parvovirus B19 (PVB19) have been identified as relevant agents of myocarditis. The different cell tropism of these viruses implicates distinct pathogenic principles, which, at present, are not completely understood. Whereas enteroviruses damage the heart primarily via direct lysis of infected myocytes, PVB19 does not infect myocytes, but endothelial cells of small intracardiac arterioles and venules, resulting in impairment of myocardial microcirculation with secondary myocyte necrosis during acute infection. Histological and immunohistological stainings combined with molecular biological approaches in EMB will help us to resolve the question of whether patients with myocarditis should be treated by specific antiviral agents or by immunosuppressive therapies.


Subject(s)
Endocardium/pathology , Endocardium/virology , Enterovirus B, Human/isolation & purification , Myocarditis/diagnosis , Myocarditis/virology , Parvovirus B19, Human/isolation & purification , Biopsy , Enterovirus B, Human/genetics , Enterovirus B, Human/pathogenicity , Humans , Immunohistochemistry , Myocarditis/pathology , Myocarditis/therapy , Parvovirus B19, Human/genetics , Parvovirus B19, Human/pathogenicity
11.
Am J Pathol ; 162(5): 1709-20, 2003 May.
Article in English | MEDLINE | ID: mdl-12707055

ABSTRACT

To gain insight into the strategies of the immune system to confer resistance against the development of chronic coxsackievirus B3 (CVB3) myocarditis we compared the course of the disease in C57BL/6 mice, beta2-microglobulin knockout (beta2m(-/-)) mice, and perforin-deficient (perforin(-/-)) mice. We found that perforin(-/-) mice as well as immunocompetent C57BL/6 mice reveal a resistant phenotype with complete elimination of the virus from the heart in the course of acute myocarditis. In contrast, myocardial CVB3 infection of beta2m(-/-) mice was characterized by a significantly higher virus load associated with a fulminant acute inflammatory response and, as a consequence of virus persistence, by the development of chronic myocarditis. Interferon-gamma secretion of stimulated spleen cells was found to be significantly delayed in beta2m(-/-) mice compared to perforin(-/-) mice and C57BL/6 control mice during acute myocarditis. In addition, generation of virus-specific IgG and neutralizing antibodies were found to be significantly decreased in beta2m(-/-) mice during acute infection. From these results we conclude that protection against the development of chronic myocarditis strongly depends on the expression of beta2m, influencing the catabolism of IgG as well as the production of protective cytokines, such as interferon-gamma. Moreover, CVB3-induced cardiac injury and prevention of chronic myocarditis was found to be unrelated to perforin-mediated cytotoxicity in our model system.


Subject(s)
Antibodies, Viral/immunology , Coxsackievirus Infections/immunology , Enterovirus/pathogenicity , Immunoglobulin G/immunology , Interferon-gamma/genetics , Myocarditis/virology , beta 2-Microglobulin/immunology , Animals , Chlorocebus aethiops , Coxsackievirus Infections/pathology , Disease Models, Animal , Gene Expression Regulation/immunology , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/immunology , Myocarditis/pathology , Perforin , Pore Forming Cytotoxic Proteins , Vero Cells , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...