• No results found

Basal and Reovirus-Induced Beta Interferon (IFN-β) and IFN-β-Stimulated Gene Expression Are Cell Type Specific in the Cardiac Protective Response

N/A
N/A
Protected

Academic year: 2019

Share "Basal and Reovirus-Induced Beta Interferon (IFN-β) and IFN-β-Stimulated Gene Expression Are Cell Type Specific in the Cardiac Protective Response"

Copied!
9
0
0

Loading.... (view fulltext now)

Full text

(1)

0022-538X/05/$08.00⫹0 doi:10.1128/JVI.79.5.2979–2987.2005

Copyright © 2005, American Society for Microbiology. All Rights Reserved.

Basal and Reovirus-Induced Beta Interferon (IFN-

) and

IFN-

-Stimulated Gene Expression Are Cell Type

Specific in the Cardiac Protective Response

Michael J. Stewart,

1

Kathleen Smoak,

1

Mary Ann Blum,

2

and Barbara Sherry

1,2

*

Department of Microbiology, College of Agriculture and Life Sciences,1and Department of

Molecular Biological Sciences, College of Veterinary Medicine,2North Carolina

State University, Raleigh, North Carolina

Received 6 May 2004/Accepted 9 October 2004

Viral myocarditis is an important human disease, with a wide variety of viruses implicated. Cardiac myocytes are not replenished yet are critical for host survival and thus may have a unique response to infection. Previously, we determined that the extent of reovirus induction of beta interferon (IFN-) and IFN--mediated protection in primary cardiac myocyte cultures was inversely correlated with the extent of reovirus-induced cardiac damage in a mouse model. Surprisingly, and in contrast, the IFN-response did not determine reovirus replication in skeletal muscle cells. Here we compared the IFN-response in cardiac myocytes to that in primary cardiac fibroblast cultures, a readily replenished cardiac cell type. We compared basal and reovirus-induced expression of IFN-, IRF-7 (an interferon-stimulated gene [ISG] that further induces IFN-), and another ISG (561) in the two cell types by using real-time reverse transcription-PCR. Basal IFN-, IRF-7, and 561 expression was higher in cardiac myocytes than in cardiac fibroblasts. Reovirus T3D induced greater expression of IFN-in cardiac myocytes than in cardiac fibroblasts but equivalent expression of IRF-7 and 561 in the two cell types (though fold induction for IRF-7 and 561 was higher in fibroblasts than in myocytes because of the differences in basal expression). Interestingly, while reovirus replicated to equivalent titers in cardiac myocytes and cardiac fibroblasts, removal of IFN-resulted in 10-fold-greater reovirus replication in the fibroblasts than in the myocytes. Together the data suggest that the IFN-response controls reovirus replication equivalently in the two cell types. In the absence of reovirus-induced IFN-, however, reovirus replicates to higher titers in cardiac fibroblasts than in cardiac myocytes, suggesting that the higher basal IFN-and ISG expression in myocytes may play an important protective role.

Viral myocarditis has occurred in an estimated 5 to 20% of the human population. It is often fatal in infants and, although usually resolved in adults, can progress to chronic myocarditis, dilated cardiomyopathy, and/or cardiac failure (62). In recent years, clinical trials of antiviral agents have shown promise in the treatment of the disease. Specifically, alpha interferon (IFN-␣) (9, 17, 36, 37, 55) and IFN-␤(26) have demonstrated an ability to decrease viral load, improve cardiac function, and increase the survival rate in patients with the disease. However, none of these therapies can completely restore cardiac function. Many viruses have been implicated in human myocarditis (62), with enteroviruses and adenoviruses accounting for the majority of cases (33, 42, 43, 58). Enterovirus-induced myocar-ditis is both immune mediated (8, 42) and due to direct cyto-pathic effect (6, 20). In human myocarditis associated with adenovirus (33) or human immunodeficiency virus, the severity of disease correlates poorly with the presence of an inflamma-tory infiltrate, suggesting a direct viral effect. However, non-immune-mediated mechanisms of viral myocarditis remain largely unexplored.

Reovirus-induced myocarditis is not immune mediated (49, 50); indeed, the immune system is protective against cardiac damage (50). Instead, reovirus-induced myocarditis reflects

vi-rally induced apoptosis of cardiac cells (11). Thus, reovirus infection provides an excellent model for studying direct cyto-pathic effect in the heart. Previously, we demonstrated that in primary cardiac myocyte cultures (PCMCs), reovirus induction of, and sensitivity to, IFN-␤is a determinant of viral myocar-ditic potential. Specifically, nonmyocarmyocar-ditic reoviruses induce more IFN-␤and/or are more sensitive to the antiviral effects of IFN-␤than myocarditic reoviruses (52).

Importantly, IFN-␤mediates its antiviral effects though the induction of IFN-stimulated genes (ISGs). Following secretion from infected cells, IFN-␤binds to the IFN-␣/␤receptor, re-sulting in receptor dimerization and activation of receptor-associated kinases of the Janus kinase family (JAKs). Specifi-cally, Jak1 and Tyk2 are activated and phosphorylate tyrosine residues on STAT1 and STAT2 (18, 39, 46, 53). These newly phosphorylated STAT proteins associate with interferon reg-ulatory factor 9 (IRF-9, or p48) to form the multimeric protein complex ISGF3, which migrates into the nucleus to bind gene promoter regions containing an IFN-stimulated response ele-ment (ISRE) (53).

To date, more than 300 genes have been identified as ISGs (5, 12, 13). Classical ISGs include those encoding PKR, 2⬘-5⬘ oligoadenylate synthetase, and Mx family GTPases. Notably, while PKR is critical for protection against reovirus-induced myocarditis (54), the role of other ISGs in protection against myocarditis is unclear. The ISG 561-encoded protein, p56, is the most strongly induced gene product following addition of

* Corresponding author. Mailing address: Department of Molecular Biological Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606. Phone: (919) 515-4480. Fax: (919) 515-3044. E-mail: barbara_sherry@ncsu.edu.

2979

on November 8, 2019 by guest

http://jvi.asm.org/

(2)

do not generate IRF-7 following IFN treatment (45). Cardiac myocytes are exposed to a variety of viruses, are not replenished, and yet are critical to survival, suggesting that this highly specialized cell type may have evolved a unique re-sponse to viral infection. We have previously shown that reo-virus induction of IFN-␤ and IFN-␤-mediated protection in PCMCs correlate inversely with viral spread in PCMCs and the degree of cardiac damage in a mouse model (52). Surprisingly, and in contrast, the IFN response in skeletal muscle cells is not a determinant of viral replication (52), suggesting a cell type-specific role for IFN-␤in antiviral protection. Here, we com-pare the IFN-␤response in PCMCs to that in primary cardiac fibroblast cultures (PCFCs), a readily replenished cardiac cell type. We use the nonmyocarditic reovirus strain T3D, because it both induces IFN-␤well and is highly sensitive to the anti-viral effects of IFN-␤, a characteristic not common to myocar-ditic reovirus strains (52). Results demonstrate that IFN-␤and the ISGs IRF-7 and 561 are each basally expressed at higher levels in PCMCs than in PCFCs. Moreover, while reovirus T3D induced expression of IFN-␤to significantly higher levels in PCMCs than in PCFCs, it induced similar expression of IRF-7 and 561 in the two cell types. Interestingly, we also show that while reovirus replicated to equivalent titers in PCMCs and PCFCs, removal of IFN-␤resulted in significantly greater reovirus replication in PCFCs than in PCMCs. Together, these data indicate that the IFN-␤response is more critical for pro-tection in PCFCs than in PCMCs. Additionally, these data suggest that in the absence of IFN-␤-stimulated gene expres-sion, higher basal IFN-␤and ISG expression in PCMCs may play a protective role.

MATERIALS AND METHODS

Mice and inoculations.Timed pregnant Cr:NIH(S) mice were obtained from the National Cancer Institute. IFN-␣/␤receptor⫺/⫺(38) and wild-type parental

129Sv/Ev mice were maintained in breeding colonies for timed matings to gen-erate fetuses for primary cultures. Mice were housed according to the recom-mendations of the Association for Assessment and Accreditation of Laboratory Animal Care, and all procedures were approved by the North Carolina State University institutional animal care and use committee.

Cell cultures.To generate primary cardiac myocyte and fibroblast cultures from Cr:NIH(S), IFN-␣/␤receptor⫺/⫺(38), or 129Sv/Ev mice, term fetuses or

1-to 2-day-old neonates were euthanized and the apical two-thirds of the hearts were removed, minced, and trypsinized (3). Cells were plated at a density of 1.25

⫻106

per well in six-well clusters (Costar, Cambridge, Mass.) and incubated for 1.5 to 2 h in order to isolate rapidly adhering cardiac fibroblasts. Fibroblasts were trypsinized from six-well clusters and resuspended in Dulbecco’s modified Eagle medium (DMEM) (Gibco BRL, Gaithersburg, Md.) supplemented with 7% fetal calf serum (HyClone, Logan, Utah) and 50␮g of Mezlin (Miles Labs) per ml. Fibroblasts were then plated as indicated for each procedure. The remaining

Infections for RNA harvests.Reovirus infected similar percentages of cells in PCMCs and PCFCs by immunocytochemistry (data not shown). PCMCs were plated at confluency (106

cells), and PCFCs were plated at half-confluency (5⫻ 105cells), in a 24-well cluster; PCFCs were confluent by the day of infection (106

cells). Two days postplating, PCMCs or PCFCs were washed twice with supple-mented DMEM and then immediately infected with reovirus T3D (Dearing) at 10 PFU per cell in 300␮l of supplemented medium. After incubation for 1 h at 37°C under 5% CO2, 700␮l of a DMEM overlay was added, and these cultures

were harvested at the indicated times. “Mock infected” refers to uninfected cultures washed with supplemented DMEM and harvested either immediately or 5 to 6 h after being overlaid with 1 ml of supplemented DMEM. For harvests performed at 0 h, virus was added and immediately removed (with no incubation time), and cells were harvested.

Infections for viral replication studies.PCMCs and PCFCs were plated at densities of 4.0⫻105and 2.0105per well, respectively, in 300l in 96-well

tissue culture plates (Costar). At 2 days postplating, cultures were infected with a panel of parental and reassortant reoviruses at 0.1 PFU per cell. Cultures were treated with either 3␮l of an antibody containing 165 National Institutes of Health neutralizing units of rabbit-anti-mouse IFN-␣/␤(catalog no. 21032; Lee Biomolecular Research, Inc., San Diego, Calif.) or a control antibody. Seven days postinfection, cell monolayers were frozen at⫺70°C and subjected to additional freeze-thaw cycles. Cultures were then lysed in 0.5% Nonidet P-40, and titers were determined by plaque assay on mouse L-cell monolayers as previously described (48).

RNA harvest and reverse transcription.At the indicated times postinfection, supernatants were aspirated. Cells were lysed directly from the culture plates and homogenized by using Qiashredders (QIAGEN), and total RNA was isolated by using an RNeasy kit (QIAGEN). Contaminating genomic DNA was removed by DNase treatment using an RNase-free DNase set (QIAGEN). Total RNA was extracted and quantified in initial experiments by fluorometry using the Ribo-Green total RNA detection kit (Molecular Probes; Eugene, Oreg.). RNA from each well was then subjected to reverse transcription using the following com-ponents (final concentrations) for a total reaction volume of 50␮l: 5␮M oli-go(dT) (Promega), 1⫻Taq buffer, 7.5 mM MgCl2, 1 mM dithiothreitol, 1 mM

each deoxynucleoside triphosphate, 0.5 U of RNA Guard (Pharmacia)/␮l, and 0.22 U of avian myeloblastosis virus reverse transcriptase (Promega)/␮l.

Real-time PCR. Approximately 10% of the reverse transcription reaction product was then amplified by real-time PCR. Experiments were performed in duplicate 25-␮l reaction mixtures in 96 well plates (Bio-Rad) using Quantitech master mix (QIAGEN) spiked with 10 nM fluorescein (Molecular Probes) to optimize fluorescent data quality and analysis. Duplicate PCRs were carried out in 96-well plates with optical sealing tape (Bio-Rad). Amplification, quantifica-tion, and melting curve analysis (detection of specific products) were performed on an iCycler iQ fluorescence thermocycler (Bio-Rad) with the following cycle profile: 95°C (PCR initial activation step, 13.5 min), followed by 50 cycles of 95°C for 10 s (denaturation) and 59°C for 60 s (annealing, synthesis, and fluorescent data collection step), followed by a melting curve protocol designed for decre-ment temperatures of 0.5°C with a starting temperature of 95°C and an ending temperature of 50°C.

The sequences for the primers were selected by using online software designed by Steve Rosen and Helen J. Skaletsky (1998) (available at http://www-genome .wi.mit.edu/cgi-bin/primer/primer3_www.cgi). Primers were checked for base complementarity by using http://www.operon.com/oligos/toolkit.php (online soft-ware provided by QIAGEN, 1996 to 2003). Primers were then designed as follows: IRF-7 forward primer, 5⬘CCCATCTTCGACTTCAGCAC 3⬘; IRF-7 reverse primer, 5⬘TGTAGTGTGGTGACCCTTGC 3⬘; 561 forward primer, 5⬘

on November 8, 2019 by guest

(3)

TGGCCGTTTCCTACAGTTT 3⬘; 561 reverse primer, 5⬘TCCTCCAAGCAA AGGACTTC 3⬘; IFN-␤forward primer, 5⬘GGAGATGACGGAGAAGATGC 3⬘; IFN-␤reverse primer, 5⬘CCCAGTGCTGGAGAAATTGT 3⬘; glyceralde-hyde-3-phosphate dehydrogenase (GAPDH) forward primer, 5⬘CAACTACAT GGTCTACATGTTC 3⬘; GAPDH reverse primer, 5⬘CTCGCTCCTGGAAGA TG 3⬘.

Note that the sequence of the IRF-7 amplicon is present in all three IRF-7 spliceoforms (␣,␤, and␥); therefore, all spliced variants are detected. To de-termine the copy number of experimental samples, standard curves of known concentrations of DNA were generated for each gene of interest on each plate, using four 10-fold dilutions of PCR-generated DNA fragments which included the amplicon sequence. To generate these DNA fragments, the following prim-ers were used to amplify sequences from mouse DNA: IRF-7 forward primer, 5⬘ TGGGTTCCTGGATGTGAC 3⬘; IRF-7 reverse primer, 5⬘TTCACCAGGAT CAGGGTC 3⬘; 561 forward primer, 5⬘ CTGAGGCCCACATTTGAGAT 3⬘; 561 reverse primer, 5⬘GGAGCATTGGAACACTTGGT; IFN-␤forward prim-er, 5⬘GCGTTCCTGCTGTGCTTC 3⬘; IFN-␤reverse primer, 5⬘CCATCCAG GCGTAGCTG 3⬘; GAPDH forward primer, 5⬘ GTGAAGGTCGGTGTGA ACGG 3⬘; GAPDH reverse primer, 5⬘GTGGCAGTGATGGCATGGAC 3⬘.

These DNA fragments were then purified by gel extraction and quantified by fluorometry. iCycler software was used to analyze data (confirming single peak melt curve) and generate standard curves on each plate, and then copy numbers for the gene of interest were determined relative to GAPDH expression. Al-though the average total yields of RNA were similar for PCMCs and PCFCs (1.3-fold difference as determined by fluorometry [data not shown]), GAPDH basal expression was 3.0-fold greater in PCMCs than in PCFCs (data not shown). Thus, data were normalized to compensate for this difference. GAPDH expres-sion remained constant, regardless of reovirus infection or addition of IFN (data not shown). Controls using water or reverse-transcribed water were included for each primer set and were always negative.

Statistical analysis.A Student one-tailedttest and pooled variance were used for statistical analysis. Data were subjected to outlier analysis (Systat 9.0). For analysis of viral replication, the nonparametric Kruskal-Wallis test (Systat 9.0) was used. In all cases, results were considered significant at aPvalue ofⱕ0.05.

RESULTS

Reovirus T3D induces greater IFN-expression in PCMCs than in PCFCs.The possibility that the innate cardiac response to viral infection is cell type specific had not previously been

explored in any viral system. Here, we compared viral induc-tion of IFN-␤gene expression in PCMCs to that in PCFCs. PCMCs and PCFCs were derived from Cr:NIH(S) (wild-type) mice. Two or 3 days postplating, cultures were mock treated or virally infected, total RNAs were extracted and reverse tran-scribed, and cDNAs were then analyzed by real-time PCR to determine IFN-␤absolute expression (mRNA copy number). Basal IFN-␤expression was 3.0-fold higher in PCMCs than in PCFCs (Fig. 1A) (P ⬍ 0.001). In addition, at multiple time points postinfection, T3D induced IFN-␤ expression (copy number) to significantly higher levels in PCMCs than in PCFCs (Fig. 1B) (3.8-fold at 4 h [P⫽0.03]; 12.8-fold at 8 h [P⫽0.01]). When results from Fig. 1A and B were combined to express data as fold induction, T3D was again found to induce IFN-␤ expression more in PCMCs than in PCFCs (Fig. 1C).

[image:3.585.65.518.70.263.2]

Basal IRF-7 expression is higher in PCMCs than in PCFCs, but reovirus induction of IRF-7 is similar.Basal and reovirus-induced expression of IFN-␤ was greater in PCMCs than in PCFCs; thus, we inquired whether basal and/or induced ISG expression was similarly higher in PCMCs. IRF-7 was specifi-cally chosen for this experiment because it is an ISG solely regulated by ISGF3 (45) and furthermore because it is critical to the robust amplification of the IFN-␤ response. Reverse transcription and real-time PCR were performed on RNAs from mock- and virus-infected PCMCs and PCFCs as de-scribed above. Basal expression of IRF-7 was dramatically higher in PCMCs than in PCFCs (12.7-fold;P⬍ 0.001 [Fig. 2A]). In contrast, reovirus T3D induced similar IRF-7 expres-sion (copy numbers) in PCMCs and PCFCs (Fig. 2B) except at 8 h postinfection, when the difference was only 1.9-fold (with no increase in difference at later times [data not shown]). When the data in Fig. 2A and B were combined, fold induction

FIG. 1. Basal and induced IFN-␤gene expression is greater in PCMCs than in PCFCs. PCMCs and PCFCs were derived from Cr:NIH(S) mice. Cultures were mock treated or infected with reovirus T3D at a multiplicity of infection of 10 PFU/cell. Total RNA was extracted and quantified via fluorometry. Equivalent RNA was reverse transcribed, cDNAs were analyzed via real-time PCR, and the IFN-␤copy number was normalized to the GAPDH copy number for each well. Asterisk indicates a significant difference in IFN-␤expression between PCMCs and PCFCs. (A) Basal expression of IFN-␤in PCMCs and PCFCs. Bars are averages of six replicates from three independently derived myocyte and fibroblast cultures ⫾standard errors of the means (SEM). (B) Reovirus T3D induction of IFN-␤in PCMCs and PCFCs. Each time point is the average of five or six replicates from three independently derived myocyte and fibroblast cultures (except for PCFC values for 2 and 4 h, which are averages of four replicates from two cultures)⫾SEM (present, but not discernible for all symbols). (C) Data from panels A and B are combined to express results as fold induction over mock expression. Note that, since these data are ratios of data from the two previous panels, there are no SEM.

on November 8, 2019 by guest

http://jvi.asm.org/

(4)

of IRF-7 was found to be dramatically higher in PCFCs (3.5- to 17.5-fold) than in PCMCs (0.63- to 2.3-fold) (Fig. 2C). Thus, dramatically different basal levels of IRF-7 expression in the two cell types resulted in a large difference in fold induction, despite a minimal difference in final copy number. Interest-ingly, when reovirus T3D was added to PCMCs and PCFCs and then immediately washed off, IRF-7 was induced in PCFCs but not in PCMCs relative to basal expression (3.8-fold in PCFCs [P⬍0.01], similar to fold induction at 2 h in Fig. 2C; 0.84-fold in PCMCs [P⬎0.1] [data not shown]). Future studies will address whether this finding reflects cell type-specific sig-naling upon virus binding.

[image:4.585.72.516.65.234.2]

Basal expression of ISG 561 is higher in PCMCs than in PCFCs, but reovirus induction of 561 is similar.The ISG 561 is the most strongly induced ISG following treatment of human cells with IFN-␣/␤ (12), but unlike induction of IRF-7, virus has been shown to directly induce expression from the 561 promoter in the absence of IFN (15, 59). This direct induction of 561 is most likely mediated by IRF-3 or IRF-1 binding to the 561 ISRE (2, 7, 10, 14, 15, 40). Importantly, direct induction of 561 is cell type and/or stimulus specific (2, 19). As with IRF-7, basal expression of 561 was higher in PCMCs than in PCFCs (3.6-fold;P⬍0.001 [Fig. 3A]). However, in contrast to IRF-7, reovirus T3D induced 561 with cell type-specific kinetics (Fig.

FIG. 2. IRF-7 gene expression in PCMCs and PCFCs. Reverse transcription and real-time PCR were performed on RNAs from mock- and T3D-infected PCMCs and PCFCs as for Fig. 1. Asterisk indicates a significant difference in IRF-7 expression between PCMCs and PCFCs. (A) Basal expression of IRF-7 in PCMCs and PCFCs. Bars are averages of 17 or 18 replicates from at least eight independently derived primary cell cultures⫾standard errors of the means (SEM). (B) Reovirus T3D induction of IRF-7 (copy number) in PCMCs and PCFCs. Each time point is the average of six to eight replicates from three to four myocyte and fibroblast cultures (except for PCFC values for 2 and 4 h, which are averages of four replicates from two cultures)⫾SEM. (C) Data from panels A and B are combined to express results as fold induction over mock expression. Again, because these data are expressed as ratios of data from the two previous panels, there are no SEM.

FIG. 3. 561 gene expression in PCMCs and PCFCs. Reverse transcription and real-time PCR were performed on RNAs from mock- and T3D-infected PCMCs and PCFCs as for Fig. 1. Asterisk indicates a significant difference in 561 expression between PCMCs and PCFCs. (A) Basal expression of 561 in PCMCs and PCFCs. Bars are averages of 20 or 16 replicates (respectively) from at least eight independently derived primary cell cultures⫾standard errors of the means (SEM). (B) Reovirus T3D induction of 561 (copy number) in PCMCs and PCFCs. Each time point is the average of 6 to 10 replicates from three to five myocyte and fibroblast culturesSEM. (C) Data from panels A and B are combined to express results as fold induction over mock expression. Again, because these data are expressed as ratios of data from the two previous panels, there are no SEM.

on November 8, 2019 by guest

[image:4.585.67.525.485.658.2]
(5)

3B). After infection, the copy number of 561 increased and then decreased after 4 h in PCFCs but continued to rise in PCMCs, resulting in a significant difference in 561 expression at 8 h postinfection in PCMCs (fourfold;P⬍0.001). Expres-sion of 561 remained elevated in PCMCs at 10 and 12 h postinfection (data not shown). As with IRF-7, when the data in Fig. 3A and B were combined, fold induction provided a different curve because of the differences in basal expression. At 4 h postinfection, despite little difference in absolute copy numbers, fold induction differed dramatically: 12.1-fold in PCMCs compared to 52.7-fold in PCFCs (Fig. 3C). Moreover, while at 8 h postinfection PCMCs expressed fourfold more 561 than did PCFCs (Fig. 3B), there was a negligible difference in relative induction of 561 between the cell types (Fig. 3C). Thus, as with IRF-7, differences in basal 561 expression between PCMCs and PCFCs resulted in a curve for fold induction that did not reflect the actual final copy numbers.

IFN-/treatment is similar to viral infection. To deter-mine whether viral induction of IRF-7 and 561 in PCMCs and PCFCs reflected additional virus-specific events other than induction of IFN-␤, PCMCs and PCFCs were overlaid with a medium containing IFN-␣/␤and then harvested for analysis by real-time PCR. While IFN-␤induced the expression of IRF-7 (Fig. 4A) more rapidly than T3D did (Fig. 2B), the overall pattern of induction was similar, in that copy numbers of IRF-7 were comparable in PCMCs and PCFCs but fold induction (Fig. 4B) was dramatically different due to differences in basal expression. In contrast, while IFN-␣/␤ induction of the ISG 561 in PCFCs (Fig. 5A) followed kinetics similar to those of reovirus induction of 561 in that cell type (Fig. 3B), IFN-␣/␤ (Fig. 5A) and reovirus (Fig. 3B) induction of 561 followed different kinetics in PCMCs. Specifically, while 561 expression remained elevated following reovirus infection, 561 expression rapidly decreased in PCMCs following IFN-␣/␤ stimulation,

FIG. 4. (A) IFN-␣/␤induction of IRF-7 in PCMCs and PCFCs. Reverse transcription and real-time PCR were performed on RNAs from mock-and IFN-treated PCMCs mock-and PCFCs as for Fig. 1. Asterisk indicates a significant difference in IRF-7 expression between PCMCs mock-and PCFCs. Each time point is the average of four to eight replicates from two or four myocyte and fibroblast cultures⫾standard errors of the means (SEM). (B) Data from Fig. 2A and 4A are combined to express results as fold induction over mock expression. Again, because these data are expressed as ratios of data from the two previous panels, there are no SEM.

FIG. 5. (A) IFN-/induction of 561 in PCMCs and PCFCs. Reverse transcription and real-time PCR were performed on RNAs from mock-and IFN-treated PCMCs mock-and PCFCs as for Fig. 1. Each time point is the average of four to six replicates from two to three myocyte mock-and fibroblast cultures (except at 4 h, where values are averages of two replicates from a single experiment)standard errors of the means (SEM). (B) Data from Fig. 3A and 5A are combined to express results as fold induction over mock expression. Again, because these data are expressed as ratios of data from the two previous panels, there are no SEM.

on November 8, 2019 by guest

http://jvi.asm.org/

[image:5.585.115.470.70.223.2] [image:5.585.108.477.517.678.2]
(6)

kinetics similar to those of 561 in PCFCs. Again, differences in the fold induction of 561 expression (Fig. 5B) between PCMCs and PCFCs were largely determined by differences in basal expression.

PCFCs are more dependent on the IFN response than are PCMCs.The data presented in Fig. 1 to 5 suggest that relative (fold) induction of ISGs is determined, at least in part, by basal ISG expression. Specifically, PCMCs may rely on high basal ISG levels, thereby eliminating the need for extensive induc-tion of ISGs, while in contrast, PCFCs express low basal ISG levels but achieve protective ISG levels through rapid and substantial IFN-mediated induction. These data suggest that PCFCs may rely more than PCMCs on the IFN-mediated response for antiviral protection.

To address this possibility, PCMCs and PCFCs were in-fected with a large panel of reoviruses in the presence of control or anti-IFN antibodies. After incubation to allow viral replication and spread, cultures were harvested and viral titers were determined. In the presence of the control antibody, reovirus replicated to similar titers in PCMCs and PCFCs (Fig. 6A) and, as previously observed, myocarditic viruses replicated to higher titers than nonmyocarditic viruses in cardiac cells (52). However, in the presence of an anti-IFN antibody, reo-virus replicated to significantly higher titers in PCFCs than in PCMCs. Specifically, the anti-IFN antibody increased viral rep-lication an average of 50-fold in PCMCs but 550-fold in PCFCs (Fig. 6B) (P⬍0.001). These results demonstrate that induced IFN provides greater antiviral protection in PCFCs than in PCMCs. The viral genes that determined the degree of IFN-mediated protection in PCFCs (data not shown) were the same as those previously identified for PCMCs (52). Given that viruses replicated equivalently in PCMCs and PCFCs in con-trol cultures but that in the absence of IFN, viruses replicated to titers 10-fold higher in PCFCs than in PCMCs, the data suggest that constitutively higher ISG levels in PCMCs may provide a degree of protection even in the absence of IFN.

Viruses can directly induce ISGs such as 561 (15, 59), and the mechanisms may be cell type specific. That is, it is possible that the residual protection in the absence of IFN in PCMCs relative to PCFCs reflected greater direct induction of ISGs in

PCMCs than in PCFCs. Accordingly, we tested whether reo-virus can directly induce expression of 561 in PCMCs and PCFCs derived from IFN-␣/␤receptor-null (IFN-R-null) mice. PCMCs and PCFCs were derived from IFN-R-null mice and were infected with reovirus T3D. Cultures were harvested at 0 and 8 h postinfection. By real-time PCR analysis, reovirus directly induced 561 expression in both PCMCs and PCFCs, with no significant difference between cell types (Fig. 7) (P⫽ 0.11). Additionally, reovirus T3D did not induce IRF-7 expres-sion in PCMCs derived from IFN-R-null mice (data not shown). Therefore, differences in viral replication between PCFCs and PCMCs in the absence of IFN-␣/␤are not associ-ated with direct viral induction of 561 or IRF-7.

What determines the greater basal ISG expression in PCMCs relative to PCFCs? PCMCs express higher basal IFN-␤levels than do PCFCs (Fig. 1A), providing a possible explanation for

[image:6.585.77.510.66.226.2]

FIG. 6. PCFCs are more dependent on the IFN response than PCMCs. Replicate wells of PCMCs and PCFCs were infected at a multiplicity of infection of 0.1 and overlaid with an anti-IFN-␣/␤antibody or a control antibody. At 7 days postinfection, cultures were lysed and viral titers were determined by a plaque assay. (A) Results expressed as mean viral yield of duplicate wells⫾standard deviation. (B) Results from PCMCs and PCFCs expressed as ratio of viral yield in anti-IFN-␣/␤-treated wells relative to that in control-treated wells.

FIG. 7. Reovirus directly induces 561 expression to similar extents in PCMCs and PCFCs. PCMCs and PCFCs were derived from IFN-R-null mice. Cultures were infected with reovirus T3D at a multiplicity of infection of 10 and were harvested at 0 and 8 h postinfection. Total RNA was extracted and reverse transcribed. Resultant cDNAs were analyzed by real-time PCR. Each bar is the average of 10 replicates from five experiments with PCMCs, or of 6 replicates from three experiments with PCFCs,the standard error of the mean. Asterisk indicates a significant difference in 561 expression between 0 and 8 h postinfection in a given cell type.

on November 8, 2019 by guest

(7)

differences between PCMCs and PCFCs in IRF-7 expression (Fig. 2A) and 561 expression (Fig. 3A). Indeed, IFN-R-null PCMCs and PCFCs express dramatically lower basal levels of 561 (Fig. 8A) and IRF-7 (Fig. 8B) than wild-type Cr:NIH(S) cultures (similar results were obtained for mock-treated paren-tal 129Sv/Ev PCMCs and PCFCs [data not shown]). Together, these data strongly suggest that basal expression of IFN is a primary determinant of basal ISG expression and that basal ISG expression is an important determinant of antiviral pro-tection in PCMCs but not PCFCs.

DISCUSSION

Cardiac myocytes are not replenished yet are critical for survival. Given that many viruses gain access to the heart, these highly specialized cells may have a unique response to viral infection. Specifically, cardiac myocytes may be more depen-dent on the innate immune response than cell types that are easily replenished. Indeed, we have previously shown that the innate IFN response is critical in determining viral spread in PCMCs but not in differentiated skeletal muscle cells (52).

In this report, we demonstrate that PCMCs express greater levels of basal IFN-␤than PCFCs (Fig. 1A). In addition, reo-virus induces greater IFN-␤ expression in PCMCs than in PCFCs, as indicated by both copy number and relative fold induction (Fig. 1A and B). Cells “primed” with small amounts of IFN-␣/␤ and then virally infected express more IFN-␣/␤ than nonprimed cells (44). This priming is IRF-7 dependent: following viral infection, spleen cells lacking IRF-3, but primed by IFN to express IRF-7, induce IFN-␣/␤ mRNA to levels similar to those of wild-type spleen cells (57). Given that basal IRF-7 expression was significantly higher in PCMCs than in PCFCs (12-fold), our data support a mechanism whereby rel-atively high basal IFN-␤expression in PCMCs results in sim-ilarly high basal IRF-7 expression, which “primes” PCMCs to induce greater IFN-␤expression in response to viral infection. This would also be in agreement with previous work suggesting that higher basal IRF-7 expression in plasmacytoid dendritic cells (PDCs) compared to monocyte-derived dendritic cells (MDDCs) results in Sendai virus inducing greater expression

and a broader set of IFN-␣ genes in PDCs than in MDDCs (24). Interestingly, basal expression of IRF-7 is higher in lym-phoid than in nonlymlym-phoid cells (1, 63). While this likely re-flects the prominent role of IRF-7 in the induction of distinct IFN-␣ genes (28), the higher basal expression of IRF-7 in PCMCs than in PCFCs could reflect a “prearming” of PCMCs, thereby allowing these cells to respond more quickly to viral infection. This could be critical for effective protection of these differentiated, nondividing, essential cells. Surprisingly, even though reovirus induced more IFN-␤in PCMCs than PCFCs (Fig. 1B), reovirus induction of IRF-7 expression (copy num-ber) was similar at all time points (Fig. 2B). These data suggest that there may be an upper limit to IRF-7 expression in cardiac cells and that cell types with higher basal expression (PCMCs) are less responsive to viral infection and/or IFN-␣/␤ stimula-tion.

[image:7.585.118.468.68.228.2]

As with IRF-7, peak expression of the ISG 561 was compa-rable in PCMCs and PCFCs (Fig. 3B), but unlike IRF-7, the kinetics of viral induction of 561 were cell type specific: induc-tion of 561 was prolonged in PCMCs but transient in PCFCs. Interestingly, following IFN treatment, the kinetics and levels of 561 expression were identical for the two cell types (Fig. 5A), resembling that in PCFCs following reovirus infection and resembling transient induction of 561 mRNA by double-stranded RNA in human glioblastoma GRE cells (2). One possibility could be that cell type-specific differences in the kinetics of induction of 561 reflect differences in virally acti-vated IRF-3. That is, IRF-3 could remain actiacti-vated longer, or could be more efficient at induction of 561 expression, in PCMCs than PCFCs. However, our data demonstrating similar reovirus-induced direct induction of 561 in PCMCs and PCFCs argue against a role for IRF-3 (see below) (Fig. 8A). Alterna-tively, it is possible that differences in the kinetics of 561 ex-pression reflect cell type-specific differences in the kinetics of reovirus-induced IRF-1 expression. Indeed, in human fibro-blasts, the pattern of peak expression and then decline of the ISG 6–16 coincides with the kinetics of IFN-induced expres-sion of IRF-1 but not ISGF3 (23, 53). Finally, it is possible that differences in accumulated levels of 561 mRNA, as measured

FIG. 8. Basal IFN expression is a determinant of basal ISG expression. (A) Basal 561 expression from Fig. 3A (wild-type cultures) compared to 561 expression at 0 h from Fig. 7 (IFN-R-null cultures). (B) Basal IRF-7 expression from Fig. 2A (wild-type cultures) compared to IRF-7 expression from IFN-R-null-derived cultures at 0 h. Asterisk indicates a significant difference in 561 (A) or IRF-7 (B) expression between wild-type and IFN-R-null cultures.

on November 8, 2019 by guest

http://jvi.asm.org/

(8)

cultures likely reflects IRF-7’s greater dependence on ISGF3-mediated transcription (data not shown) (45); in contrast, 561 can also be induced by IRFs (2, 7, 10, 14, 15, 40) and thus is likely less affected by loss of ISGF3. The greater decrease in ISG expression in PCMCs than in PCFCs likely reflects the higher basal expression levels of ISGs (IRF-7 and 561) in PCMCs than in PCFCs.

Interestingly, our results in cultures derived from IFN-R-null mice demonstrate that like vesicular stomatitis virus, en-cephalomyocarditis virus, and Sendai virus (15), reovirus T3D can directly induce expression of 561 (Fig. 7). Moreover, these cultures display no statistical difference in 561 expression levels at 8 h postinfection, indicating that in the absence of JAK/ STAT signaling, reovirus induces 561 expression to similar extents in PCMCs and PCFCs. These results contrast with a report indicating that reovirus does not directly induce the expression of p56 (encoded by 561) in GRE cells (15). Both laboratories used the Dearing strain of T3 reovirus, but this does not exclude the possibility of substrain-specific differ-ences. Alternatively, direct induction of ISGs by reovirus may be cell type specific, occurring in cardiac cells but not in human glioblastoma cells.

Viruses replicated to higher titers in PCFCs than in PCMCs in the absence of induced IFN-␤(Fig. 6). Together with data demonstrating higher basal ISG expression in PCMCs (Fig. 2 to 5), this finding suggests that PCMCs benefit from a “pre-arming” while PCFCs are more dependent on induced expres-sion of IFN-␤and ISGs. Others have shown that mutant cell lines expressing high basal levels of IFN also express high basal levels of the ISGs 6–16, 9–27, and 2⬘-5⬘ oligoadenylate syn-thetase (35). Our results are also in agreement with observa-tions that in the absence of IFN-mediated protection, cells stably transfected with an IRF-1 transgene are significantly more resistant to vesicular stomatitis virus, encephalomyocar-ditis virus, and Newcastle disease virus than wild-type cells (41). Importantly, because prearming requires higher levels of basal IFN, and IFN is antiproliferative, it is likely that this mechanism of protection is restricted to nondividing cell types such as cardiac myocytes.

Finally, our results demonstrate that relative induction of gene expression can be misleading, in that final absolute ex-pression (copy number) may differ minimally while relative induction differs drastically due to differences in basal expres-sion. That is, reovirus or IFN-␤always induced equivalent or more copies of 561 and IRF-7 in PCMCs than in PCFCs (Fig. 2B, 3B, 4A, and 5A), but relative induction was always higher

561 (0.01 GAPDH-normalized copies) than wild-type cultures expressed basally (0.02 GAPDH-normalized copies). This in-dicates that relative induction data such as those obtained from reporter gene and microarray analyses should be interpreted carefully when one is comparing multiple cell types (including null and wild type). Moreover, these data further demonstrate that basal expression can be a principal determinant of induced expression.

Future experiments will continue to identify the molecular differences between PCMCs and PCFCs following viral infec-tion and/or IFN-␤treatment. Of particular interest will be the STAT-signaling cascade. Given that relative induction of ISGs is higher in PCFCs than in PCMCs, one could imagine a more robust and/or sustained STAT activation in PCFCs.

ACKNOWLEDGMENTS

We are indebted to Tim Petty for very helpful discussions, as well as to Fred Fuller and Matthew Breen for technical assistance.

This work was supported by NHLBI grant HL57161.

REFERENCES

1.Au, W. C., P. A. Moore, D. W. LaFleur, B. Tombal, and P. M. Pitha.1998. Characterization of the interferon regulatory factor-7 and its potential role in the transcription activation of interferon A genes. J. Biol. Chem.273: 29210–29217.

2.Bandyopadhyay, S. K., G. T. Leonard, Jr., T. Bandyopadhyay, G. R. Stark, and G. C. Sen.1995. Transcriptional induction by double-stranded RNA is mediated by interferon-stimulated response elements without activation of interferon-stimulated gene factor 3. J. Biol. Chem.270:19624–19629. 3.Baty, C. J., and B. Sherry.1993. Cytopathogenic effect in cardiac myocytes

but not in cardiac fibroblasts is correlated with reovirus-induced acute myo-carditis. J. Virol.67:6295–6298.

4.Chaudhuri, J., D. Chowdhury, and U. Maitra.1999. Distinct functions of eukaryotic translation initiation factors eIF1A and eIF3 in the formation of the 40S ribosomal preinitiation complex. J. Biol. Chem.274:17975–17980. 5.Chawla-Sarkar, M., D. J. Lindner, Y. F. Liu, B. R. Williams, G. C. Sen, R. H.

Silverman, and E. C. Borden.2003. Apoptosis and interferons: role of interferon-stimulated genes as mediators of apoptosis. Apoptosis8:237–249. 6.Chow, L. H., K. W. Beisel, and B. M. McManus.1992. Enteroviral infection of mice with severe combined immunodeficiency. Evidence for direct viral pathogenesis of myocardial injury. Lab. Investig.66:24–31.

7.Collins, S. E., R. S. Noyce, and K. L. Mossman.2004. Innate cellular re-sponse to virus particle entry requires IRF3 but not virus replication. J. Virol. 78:1706–1717.

8.Cook, D. N., M. A. Beck, T. M. Coffman, S. L. Kirby, J. F. Sheridan, I. B. Pragnell, and O. Smithies.1995. Requirement of MIP-1␣for an inflamma-tory response to viral infection. Science269:1583–1585.

9.Daliento, L., F. Calabrese, F. Tona, A. L. Caforio, G. Tarsia, A. Angelini, and G. Thiene.2003. Successful treatment of enterovirus-induced myocarditis with interferon-alpha. J. Heart Lung Transplant.22:214–217.

10.Daly, C., and N. C. Reich. 1993. Double-stranded RNA activates novel factors that bind to the interferon-stimulated response element. Mol. Cell. Biol.13:3756–3764.

11.DeBiasi, R., C. Edelstein, B. Sherry, and K. Tyler.2001. Calpain inhibition protects against virus-induced apoptotic myocardial injury. J. Virol.75:351– 361.

on November 8, 2019 by guest

(9)

12.Der, S. D., A. Zhou, B. R. Williams, and R. H. Silverman.1998. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc. Natl. Acad. Sci. USA95:15623–15628. 13.de Veer, M. J., M. Holko, M. Frevel, E. Walker, S. Der, J. M. Paranjape,

R. H. Silverman, and B. R. Williams.2001. Functional classification of interferon-stimulated genes identified using microarrays. J. Leukoc. Biol. 69:912–920.

14.Grandvaux, N., M. J. Servant, B. tenOever, G. C. Sen, S. Balachandran, G. N. Barber, R. Lin, and J. Hiscott.2002. Transcriptional profiling of interferon regulatory factor 3 target genes: direct involvement in the regu-lation of interferon-stimulated genes. J. Virol.76:5532–5539.

15.Guo, J., K. L. Peters, and G. C. Sen.2000. Induction of the human protein P56 by interferon, double-stranded RNA, or virus infection. Virology267: 209–219.

16.Hamamori, Y., M. Yokoyama, M. Yamada, H. Akita, K. Goshima, and H. Fukuzaki.1990. 5-Hydroxytryptamine induces phospholipase C-mediated hydrolysis of phosphoinositides through 5-hydroxytryptamine-2 receptors in cultured fetal mouse ventricular myocytes. Circ. Res.66:1474–1483. 17.Heim, A., M. Stille-Siegner, R. Kandolf, H. Kreuzer, and H. R. Figulla.1994.

Enterovirus-induced myocarditis: hemodynamic deterioration with immuno-suppressive therapy and successful application of interferon-alpha. Clin. Cardiol.17:563–565.

18.Heim, M. H.1999. The Jak-STAT pathway: cytokine signalling from the receptor to the nucleus. J. Recept. Signal Transduct. Res.19:75–120. 19.Hershey, J. W. B.1991. Translation control in mammalian cells. Annu. Rev.

Biochem.60:717–755.

20.Herzum, M., V. Ruppert, B. Kuytz, H. Jomaa, I. Nakamura, and B. Maisch. 1994. Coxsackievirus B3 infection leads to cell death of cardiac myocytes. J. Mol. Cell. Cardiol.26:907–913.

21.Huber, S. A., and P. A. Lodge. 1984. Coxsackievirus B-3 myocarditis in BALB/c mice. Evidence for autoimmunity to myocyte antigens. Am. J. Pathol.116:21–29.

22.Hui, D. J., C. R. Bhasker, W. C. Merrick, and G. C. Sen. 2003. Viral stress-inducible protein p56 inhibits translation by blocking the interaction of eIF3 with the ternary complex eIF2.GTP.Met-tRNAi. J. Biol. Chem.278: 39477–39482.

23.Imam, A. M., A. M. Ackrill, T. C. Dale, I. M. Kerr, and G. R. Stark.1990. Transcription factors induced by interferons alpha and gamma. Nucleic Acids Res.18:6573–6580.

24.Izaguirre, A., B. J. Barnes, S. Amrute, W. S. Yeow, N. Megjugorac, J. Dai, D. Feng, E. Chung, P. M. Pitha, and P. Fitzgerald-Bocarsly.2003. Comparative analysis of IRF and IFN-␣expression in human plasmacytoid and monocyte-derived dendritic cells. J. Leukoc. Biol.74:1125–1138.

25.Kim, T. K., T. H. Kim, and T. Maniatis.1998. Efficient recruitment of TFIIB and CBP-RNA polymerase II holoenzyme by an interferon-␤enhanceosome in vitro. Proc. Natl. Acad. Sci. USA95:12191–12196.

26.Kuhl, U., M. Pauschinger, P. L. Schwimmbeck, B. Seeberg, C. Lober, M. Noutsias, W. Poller, and H. P. Schultheiss.2003. Interferon-beta treatment eliminates cardiotropic viruses and improves left ventricular function in patients with myocardial persistence of viral genomes and left ventricular dysfunction. Circulation107:2793–2798.

27.Lamphear, B. J., R. Kirchweger, T. Skern, and R. E. Rhoads.1995. Mapping of functional domains in eukaryotic protein synthesis initiation factor 4G (eIF4G) with picornaviral proteases. Implications for cap-dependent and cap-independent translational initiation. J. Biol. Chem.270:21975–21983. 28.Lin, R., P. Genin, Y. Mamane, and J. Hiscott.2000. Selective DNA binding

and association with the CREB binding protein coactivator contribute to differential activation of alpha/beta interferon genes by interferon regulatory factors 3 and 7. Mol. Cell. Biol.20:6342–6353.

29.Lin, R., C. Heylbroeck, P. M. Pitha, and J. Hiscott.1998. Virus-dependent phosphorylation of the IRF-3 transcription factor regulates nuclear translo-cation, transactivation potential, and proteasome-mediated degradation. Mol. Cell. Biol.18:2986–2996.

30.Lin, R., Y. Mamane, and J. Hiscott.1999. Structural and functional analysis of interferon regulatory factor 3: localization of the transactivating and autoinhibitory domains. Mol. Cell. Biol.19:2465–2474.

31.Lu, R., W. C. Au, W. S. Yeow, N. Hageman, and P. M. Pitha.2000. Regu-lation of the promoter activity of interferon regulatory factor-7 gene. Acti-vation by interferon and silencing by hypermethylation. J. Biol. Chem.275: 31805–31812.

32.Marie, I., J. Durbin, and D. E. Levy.1998. Differential viral induction of distinct interferon-alpha genes by positive feedback through interferon reg-ulatory factor-7. EMBO J.17:6660–6669.

33.Martin, A., S. Webber, F. Fricker, R. Jaffe, G. Demmler, D. Kearney, Y.-H. Zhang, J. Bodurtha, B. Gelb, J. Ni, T. Bricker, and J. A. Towbin.1994. Acute myocarditis: rapid diagnosis by PCR in children. Circulation90:330–339. 34.McDermott, P., M. Daood, and I. Klein.1985. Measurement of myosin

adenosine triphosphatase and myosin content in cultured heart cells. Arch. Biochem. Biophys.240:312–318.

35.McKendry, R., S. Pellegrini, I. M. Kerr, and G. R. Stark.1994. Constitutive

production of alpha and beta interferons in mutant human cell lines. J. Virol. 68:4057–4062.

36.Miric, M., A. Miskovic, J. D. Vasiljevic, N. Keserovic, and M. Pesic.1995. Interferon and thymic hormones in the therapy of human myocarditis and idiopathic dilated cardiomyopathy. Eur. Heart J.16(Suppl. O):150–152. 37.Miric, M., J. Vasiljevic, M. Bojic, Z. Popovic, N. Keserovic, and M. Pesic.

1996. Long-term follow up of patients with dilated heart muscle disease treated with human leucocytic interferon alpha or thymic hormones. Initial results. Heart75:596–601.

38.Muller, U., U. Steinhoff, L. F. L. Reis, S. Hemmi, J. Pavlovic, R. M. Zink-ernagel, and M. Aguet.1994. Functional role of type I and type II interferons in antiviral defense. Science264:1918–1921.

39.Pellegrini, S., and I. Dusanter-Fourt.1997. The structure, regulation and function of the Janus kinases (JAKs) and the signal transducers and activa-tors of transcription (STATs). Eur. J. Biochem.248:615–633.

40.Peters, K. L., H. L. Smith, G. R. Stark, and G. C. Sen. 2002. IRF-3-dependent, NF-␬B- and JNK-independent activation of the 561 and IFN-␤

genes in response to double-stranded RNA. Proc. Natl. Acad. Sci. USA 99:6322–6327.

41.Pine, R.1992. Constitutive expression of an ISGF2/IRF1 transgene leads to interferon-independent activation of interferon-inducible genes and resis-tance to viral infection. J. Virol.66:4470–4478.

42.Rose, N. R., and S. L. Hill.1996. The pathogenesis of postinfectious myo-carditis. Clin. Immunol. Immunopathol.80:S92–S99.

43.Rose, N. R., D. A. Neumann, and A. Herskowitz.1992. Coxsackievirus myo-carditis. Adv. Intern. Med.37:411–429.

44.Sato, M., S. Suemori, N. Hata, M. Asagiri, K. Ogasawara, K. Nakao, T. Nakaya, M. Katsuki, S. Noguchi, N. Tanaka, and T. Taniguchi.2000. Dis-tinct and essential roles of transcription factors IRF-3 and IRF-7 in response to viruses for IFN-␣/␤gene induction. Immunity13:539–548.

45.Sato, M., N. Tanaka, N. Hata, E. Oda, and T. Taniguchi.1998. Involvement of the IRF family transcription factor IRF-3 in virus-induced activation of the IFN-␤gene. FEBS Lett.452:112–116.

46.Schindler, C., and J. E. Darnell, Jr.1995. Transcriptional responses to polypeptide ligands: the JAK-STAT pathway. Annu. Rev. Biochem.64:621– 651.

47.Sherry, B.1998. Reoviruses II: cytopathogenicity and pathogenesis. Curr. Top. Microbiol. Immunol.233:51–63.

48.Sherry, B., C. J. Baty, and M. A. Blum.1996. Reovirus-induced acute myo-carditis in mice correlates with viral RNA synthesis rather than generation of infectious virus in cardiac myocytes. J. Virol.70:6709–6715.

49.Sherry, B., and B. N. Fields.1989. The reovirus M1 gene, encoding a viral core protein, is associated with the myocarditic phenotype of a reovirus variant. J. Virol.63:4850–4856.

50.Sherry, B., X. Li, K. L. Tyler, J. M. Cullen, and H. W. Virgin.1993. Lym-phocytes protect against and are not required for reovirus-induced myocar-ditis. J. Virol.67:6119–6124.

51.Sherry, B., F. J. Schoen, E. Wenske, and B. N. Fields.1989. Derivation and characterization of an efficiently myocarditic reovirus variant. J. Virol.63: 4840–4849.

52.Sherry, B., J. Torres, and M. A. Blum.1998. Reovirus induction of and sensitivity to beta-interferon in cardiac myocyte cultures correlate with in-duction of myocarditis and are determined by viral core proteins. J. Virol. 72:1314–1323.

53.Stark, G. R., I. M. Kerr, B. R. Williams, R. H. Silverman, and R. D. Schreiber.1998. How cells respond to interferons. Annu. Rev. Biochem. 67:227–264.

54.Stewart, M. J., M. A. Blum, and B. Sherry.2003. PKR’s protective role in viral myocarditis. Virology314:92–100.

55.Stille-Sieggener, M., A. Heim, and H. R. Figulla.1995. Subclassification of dilated cardiomyopathy and interferon treatment. Eur. Heart J.16:147–149. 56.Taniguchi, T., K. Ogasawara, A. Takaoka, and N. Tanaka.2001. IRF family of transcription factors as regulators of host defense. Annu. Rev. Immunol. 19:623–655.

57.Taniguchi, T., and A. Takaoka.2001. A weak signal for strong responses: interferon-alpha/beta revisited. Nat. Rev. Mol. Cell Biol.2:378–386. 58.Tracy, S., V. Wiegand, B. McManus, C. Gauntt, M. Pallansch, M. Beck, and

N. Chapman.1990. Molecular approaches to enteroviral diagnosis in idio-pathic cardiomyopathy and myocarditis. J. Am. Coll. Cardiol.15:1688–1694. 59.Wathelet, M. G., P. M. Berr, and G. A. Huez.1992. Regulation of gene expression by cytokines and virus in human cells lacking the type-I interferon locus. Eur. J. Biochem.206:901–910.

60.Weaver, B. K., K. P. Kumar, and N. C. Reich.1998. Interferon regulatory factor 3 and CREB-binding protein/p300 are subunits of double-stranded RNA-activated transcription factor DRAF1. Mol. Cell. Biol.18:1359–1368. 61.Wilusz, C. J., M. Wormington, and S. W. Peltz.2001. The cap-to-tail guide

to mRNA turnover. Nat. Rev. Mol. Cell Biol.2:237–246.

62.Woodruff, J. F.1980. Viral myocarditis, a review. Am. J. Pathol.101:427–479. 63.Zhang, L., and J. S. Pagano.1997. IRF-7, a new interferon regulatory factor associated with Epstein-Barr virus latency. Mol. Cell. Biol.17:5748–5757.

on November 8, 2019 by guest

http://jvi.asm.org/

Figure

FIG. 1. Basal and induced IFN-�Cultures were mock treated or infected with reovirus T3D at a multiplicity of infection of 10 PFU/cell
FIG. 2. IRF-7 gene expression in PCMCs and PCFCs. Reverse transcription and real-time PCR were performed on RNAs from mock- andT3D-infected PCMCs and PCFCs as for Fig
FIG. 4. (A) IFN-�and IFN-treated PCMCs and PCFCs as for Fig. 1. Asterisk indicates a significant difference in IRF-7 expression between PCMCs and PCFCs
FIG. 7. Reovirus directly induces 561 expression to similar extentsin PCMCs and PCFCs
+2

References

Related documents

International Journal of Scientific Research in Computer Science, Engineering and Information Technology CSEIT195335 | Received 01 May 2019 | Accepted 17 May 2019 | May June 2019 [ 5 (3)

The relative copy number of reverse transcripts in cells newly infected with wild-type, ⌬ vif, ⌬ vpr, or ⌬ vif ⌬ vpr that had been generated in 293T cells cotransfected with

We were puzzled by the punctuated staining of Gag proteins of the type C human T-cell leukemia virus type 1 (HTLV-1) that we observed both in cells transfected with the XMT

External trade statistics showing trade in agricultural products are heavily used in a number of fields, not just at the level of market divi- sions but also with regard to

It may therefore be of interest, in addition to discussing the onus of obesity, to identify and debate who the “economic problem of obesity” is of concern or interest to, and also

their respective infecting λ -like phages show relatively high similarities at amino acid sequence level, we aligned the amino acid sequence of major gene prod- ucts with known

Coreference in Knowledge Editing Coreference in Knowledge Editing K e e s v a n D e e m t e r a n d R i c h a r d P o w e r I n f o r m a t i o n T e c h n o l o g y R e s e a r c h I n s

The E6/E7 early promoter (P105) of genital human papillomavirus type 18 contains binding sites for the viral regulator E2, tandemly repeated and closely flanked by two crucial