Acute viral myocarditis is an important human disease. While enteroviruses cause immune-mediated damage, the mechanisms by which other viruses induce myocarditis remain largely unexplored. Reovirus- induced murine myocarditis is not determined by immune cell function, and presents an ideal model for studying nonimmune mediated myocarditis. Our data suggest that differential induction of and sensitivity to interferon-beta (IFN-beta) in cardiac myocytes are determinants of reovirus-induced myocarditis, and that these activities are determined by reovirus core proteins involved in RNA synthesis. IFN provides a critical first line of defense against viral infection. Viral RNA synthesis stimulates cell factors which regulate IFN-beta transcription. Secreted IFN binds receptors and induces IFN-responsive genes which mediate IFN's antiviral effects, but several of these require double stranded-[ds]RNA activation. Both induction of and sensitivity to IFN- beta are highly dependent on virus strain and cell type, and yet, there have been no studies on the cardiac IFN response to viruses. Our first hypothesis is that differential induction of IFN-beta by reovirus infection is determined by differential induction or activation of PKR, IRF-1, and/or IRF-2.
In Specific Aim 1, we will determine whether PKR (dsRNA activated protein kinase), IRF-1 (IFN regulatory factor-1) and/or IRF-2, each of which regulate IFN-beta induction in other virus/cell systems, regulate reovirus induction of IFN-beta in primary cardiac myocyte cultures, and we will use knockout mice to investigate their roles in vivo.
In Specific Aim 2, we will use two additional cell types to identify cell-specific responses. Our second hypothesis is that differential sensitivity to IFN-alpha/beta is due to differential activation of and/or sensitivity to PKR, 2',5'-oligoadenylate synthetase, and/or RNase L.
In Specific Aim 3, we will determine whether these three factors, each of which requires dsRNA activation for antiviral effects in other virus/cell systems, mediate reovirus sensitivity to IFN-alpha/beta in primary cardiac myocyte cultures, and we will use knockout mice to investigate their roles in vivo.
In Specific Aim 4, we will use two additional cell types, as above. Reoviruses offer a unique tool to investigate the IFN response invoked by closely related viruses in the heart. Our work will be the first to identity cell factors that are determinants of this cardiac IFN response and myocarditis.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL057161-01A2
Application #
2693348
Study Section
Experimental Virology Study Section (EVR)
Project Start
1998-08-07
Project End
2002-06-30
Budget Start
1998-08-07
Budget End
1999-07-31
Support Year
1
Fiscal Year
1998
Total Cost
Indirect Cost
Name
North Carolina State University Raleigh
Department
Microbiology/Immun/Virology
Type
Schools of Veterinary Medicine
DUNS #
City
Raleigh
State
NC
Country
United States
Zip Code
27695
Stewart, Michael J; Smoak, Kathleen; Blum, Mary Ann et al. (2005) Basal and reovirus-induced beta interferon (IFN-beta) and IFN-beta-stimulated gene expression are cell type specific in the cardiac protective response. J Virol 79:2979-87
Stewart, Michael J; Blum, Mary Ann; Sherry, Barbara (2003) PKR's protective role in viral myocarditis. Virology 314:92-100
Sherry, Barbara (2002) The role of interferon regulatory factors in the cardiac response to viral infection. Viral Immunol 15:17-28
Azzam-Smoak, Kathleen; Noah, Diana L; Stewart, Michael J et al. (2002) Interferon regulatory factor-1, interferon-beta, and reovirus-induced myocarditis. Virology 298:20-9
DeBiasi, R L; Edelstein, C L; Sherry, B et al. (2001) Calpain inhibition protects against virus-induced apoptotic myocardial injury. J Virol 75:351-61
Noah, D L; Blum, M A; Sherry, B (1999) Interferon regulatory factor 3 is required for viral induction of beta interferon in primary cardiac myocyte cultures. J Virol 73:10208-13