You are on page 1of 7

JOURNAL OF VIROLOGY, Sept. 2009, p. 8463–8469 Vol. 83, No.

17
0022-538X/09/$08.00⫹0 doi:10.1128/JVI.00751-09
Copyright © 2009, American Society for Microbiology. All Rights Reserved.

Inhibition of Intrahepatic Gamma Interferon Production by Hepatitis C


Virus Nonstructural Protein 5A in Transgenic Mice䌤
Tatsuo Kanda,1† Robert Steele,1 Ranjit Ray,2,3 and Ratna B. Ray1,2,3*
Departments of Pathology1 and Internal Medicine2 and Liver Center,3 Saint Louis University, St. Louis, Missouri
Received 12 April 2009/Accepted 18 June 2009

Hepatitis C virus (HCV) utilizes strategies to suppress or evade the host immune response for establishment
of persistent infection. We have shown previously that HCV nonstructural protein 5A (NS5A) impairs tumor
necrosis factor alpha (TNF-␣)-mediated apoptosis. In this study, we have examined the immunomodulatory
role of HCV NS5A protein in transgenic mouse (NS5A-Tg) liver when mice were challenged with an unrelated
hepatotropic adenovirus as a nonspecific stimulus. Hepatotropic adenovirus was introduced intravenously into
NS5A-Tg mice and control mice, and virus clearance from liver was compared over a time course of 3 weeks.

Downloaded from http://jvi.asm.org/ on May 24, 2017 by guest


The differential mRNA expression levels of 84 cytokine-related genes, signal pathway molecules, transcription
factors, and cell surface molecules were determined using real-time reverse transcription-PCR array. NS5A-Tg
mice failed to clear adenovirus from liver up to 3 weeks postinfection while control mice cleared virus within
1 to 2 weeks. Subsequent study revealed that gamma interferon (IFN-␥) expression is inhibited at both the
mRNA and protein levels in NS5A-Tg mice, and an inverse expression of transcription factors Gata-3 and
Tbx21 is observed. However, TNF-␣ mRNA and protein expression were elevated in both NS5A-Tg and control
mice. Together, our results suggested that HCV NS5A acts as an immunomodulator by inhibiting IFN-␥
production and may play an important role toward establishment of chronic HCV infection.

Hepatitis C virus (HCV), a member of Flaviviridae, causes transient viral clearance in the context of an intrahepatic HCV-
chronic infection associated with cirrhosis and hepatocellular specific T-cell response (26, 41, 47, 52). An increase in the
carcinoma. The HCV genome containing positive-strand RNA IFN-␥ gene was not detected by microarray or by quantitative
is approximately 9.6 kb and encodes a polyprotein precursor of PCR in the liver of chimpanzees chronically infected with
about 3,000 amino acids, which is cleaved by both viral and HCV compared with HCV-uninfected chimpanzees (4, 16).
host proteases into structural (core, E1, E2, and p7) and non- IFN-␥ mRNA expression levels in peripheral blood mononu-
structural (NS2, NS3, NS4A, NS4B, NS5A, and NS5B) pro- clear cells of patients are significantly lower in nonresponders
teins. HCV NS5A exists as two phosphoproteins, p56 and p58, (15). During the first 6 months of onset of HCV infection, the
phosphorylated at serine residues after the mature protein is number of IFN-␥-producing HCV-specific CD8⫹ T cells is
released from the polyprotein (46). HCV NS5A has an anti- associated with virus eradication (13). IFN-␥ also inhibits HCV
apoptotic effect (11, 27) and modulates immune responses genome replication in vitro (9). On the other hand, tumor
(1, 39). necrosis factor alpha (TNF-␣), a primary initiator of innate
Interferons (IFNs) are key players of the innate immune immune response, determines the magnitude and course of
response to virus infection. alpha IFN (IFN-␣) and IFN-␤ acquired immune responses. TNF-␣-induced apoptosis is im-
(type I IFNs) are secreted by almost all virus-infected cells and portant for clearance of virus-infected cells, and IFN-␥ accel-
by specialized blood lymphocytes. In contrast, the production erates the killing of virus-infected cells (17, 32, 48, 52).
of IFN-␥ (type II IFN) is restricted to cells of the immune A number of viruses have been shown to encode proteins
system, such as natural killer (NK) cells, macrophages, and T that have the potential to inhibit antiviral activity of innate and
cells. Although the antiviral effect of type I IFN on the repli- adaptive immune responses. Inflammatory cytokines contrib-
cation of HCV has been the subject of extensive research and
ute to viral clearance during acute viral hepatitis in humans,
although the endogenous type I IFN antiviral effector pathway
and the induction of these cytokines in the liver and other
is known to be broadly activated during chronic HCV disease
tissues of chronically infected patients may have therapeutic
(25), little is known about the role of other cytokines in cellular
value (20). Disson et al. (8) have shown that transgenic mice
defense against HCV. Further, how HCV manages to evade
expressing HCV polyprotein failed to clear an adenoviral in-
the host defense is poorly understood.
fection. However, transgenic mice expressing either core or
In the liver of chimpanzees acutely infected with HCV,
structural proteins do not appear to be immunologically
IFN-␥ was detected only in animals displaying sustained or
impaired as they are fully competent to eliminate hepato-
cytes infected with adenovirus (Ad) (24, 42). Together,
* Corresponding author. Mailing address: Department of Pathology, these results suggest a role of HCV nonstructural protein(s)
Saint Louis University, 1100 S. Grand Blvd., DRC-207, St. Louis, MO in the modulation of the immune system. In this present
63104. Phone: (314) 977-7822. Fax: (314) 771-3816. E-mail: rayrb@slu study, we have examined the role of HCV NS5A protein on
.edu.
† Present address: Chiba University, School of Medicine, Chiba,
intrahepatic cytokine production for clearance of an unre-
Japan. lated hepatotropic Ad using a transgenic mouse model. Our

Published ahead of print on 24 June 2009. results demonstrated that IFN-␥ production is inhibited and

8463
8464 KANDA ET AL. J. VIROL.

that Ad clearance is impaired in NS5A-transgenic (NS5A-


Tg) mouse liver.

MATERIALS AND METHODS


HCV NS5A-Tg mice. Transgenic FVB mice were generated by targeting the
HCV NS5A genomic region from genotype 1a, cloned under the control of a
mouse major urinary promoter in the hepatocytes (29). The major urinary pro-
moter is developmentally regulated and expressed in the liver after birth. These
mice express HCV NS5A in the liver, are phenotypically similar to their normal
littermates, and do not exhibit detectable histological changes in liver for 6 to 24
months. The expression of the transgene was comparable to that in HCV-
infected liver in humans (29). NS5A-Tg mice or control (nontransgenic [NTg])
mice aged 10 to 12 weeks were used in this study.
Ad infection. A recombinant replication-deficient Ad designated AdGFP was
kindly provided by Bert Vogelstein (Johns Hopkins University, Baltimore, MD).
This virus system is based on the human Ad type 5 from which the E1a and E1b
genes were deleted and is capable of infecting murine hepatocytes. Infecting cells
with AdGFP expresses green fluorescent protein (GFP) under the control of a

Downloaded from http://jvi.asm.org/ on May 24, 2017 by guest


cytomegalovirus promoter. Stocks of AdGFP were grown in 293 cells and purified by
two rounds of CsCl density gradient centrifugation as described previously (10).
Virus titers were determined by plaque assay on 293 cells, and a single stock was used
throughout this study. Mice (12 in each group and for each time point) were injected
with a single dose of AdGFP (1.0 ⫻ 109 PFU/mouse) or with 100 ␮l of saline as
control via tail vein. Infected mice were sacrificed at the time points indicated in Fig.
1. Livers were frozen with optimal cutting temperature medium for histological
examination and snap frozen for RNA or protein analysis.
GFP expression in AdGFP-infected mouse liver. Cryostat-frozen liver sections
(5 ␮m) were examined by indirect fluorescence microscopy for in vivo expression
of GFP in AdGFP-infected mouse liver (Bio-Rad 1024; Bio-Rad Laboratories,
Hercules, CA). One or two slides were examined for each liver specimen. GFP
expression was also quantitated using a fluorometer (1420 Victor Multilabel
Counter; Perkin Elmer, Wellesley, MA) as described earlier (2). We also exam-
ined GFP expression using immunoblot analysis. Proteins were subjected to
electrophoresis on a 12% polyacrylamide gel and transferred onto a nitro-
cellulose membrane (Bio-Rad). The membrane was probed with a monoclo-
nal antibody to GFP or ␤-tubulin (Santa Cruz Biotechnology, Santa Cruz,
CA). Protein bands were visualized using enhanced chemiluminescence
(Pierce, Rockford, IL).
ELISA. Mouse sera were analyzed for circulating IFN-␥ and TNF-␣ using an
enzyme-linked immunosorbent assay (ELISA; R & D System, Minneapolis, MN)
following the manufacturer’s protocol. Briefly, serum samples were incubated in
plates at 4°C overnight, followed by incubation with biotinylated monoclonal
antibodies. Avidin-conjugated peroxidase was added to the plates, and enzyme
activity was detected with an ELISA plate reader.
RNA extraction and real-time PCR. Total RNA was extracted from mouse FIG. 1. Impaired clearance of Ad from NS5A-Tg mouse liver.
liver (day 14 after Ad infection) using a Purescript RNA isolation kit (Gentra (A) NTg (panels a, c, e, and g) and NS5A-Tg mice (panels b, d, f, and
Systems, Minneapolis, MN). We isolated RNA from individual livers of six mice h) were infected with replication-deficient recombinant Ad expressing
(i.e., not pooled samples) from each group. Each mouse liver RNA sample was GFP by injection through the tail vein (1 ⫻ 109 PFU). Mice were
used in each PCR array. cDNA synthesis was performed using a random hex- sacrificed at day 3 (a and b), day 7 (c and d), day 14 (e and f), or day
amer. Comparison of the relative expression levels of 84 inflammatory cytokines 21 (g and h), and liver sections were evaluated microscopically for GFP
and T-cell marker-related genes was performed with a mouse PCR array (Su- expression. (B) GFP expression was quantitated from the crude liver
perarray Bioscience Corporation, Frederick, MD). For RNA quantitation, real- extracts of NS5A-Tg and NTg mice at different days. GFP expression
time PCR was conducted using SYBR Green I (ABI Prism 7700; Applied from day 3 postinfection mouse liver was chosen arbitrarily as 100%.
Biosystems, Foster City, CA). The two housekeeping genes (glyceraldehyde-3- (C) GFP expression from liver extracts (day 21) of NS5A-Tg and NTg
phosphate dehydrogenase and actin) were used for normalization, and data were mice was analyzed using specific antibody. The blot was reprobed with
analyzed by the comparative threshold cycle (CT) method. We have done the an antibody to ␤-tubulin for comparison of protein load.
assay on day 14 from each group following Ad infection. Relative quantification
of gene expression using the 2⫺⌬⌬Ct method correlated with the absolute gene
quantification obtained using a standard curve (40). Data were analyzed with
RT2 profiler PCR array data analysis software (http://www.superarray.com
and NTg mice (12 in each group) were injected with ⬃1 ⫻ 109
/pcrarraydataanalysis.php). Genes were annotated using Mouse Genome Infor-
matics (http://www.informatics.jax.org/). The relative differences were calculated PFU of AdGFP via tail vain. Livers were collected from mice
by comparing untreated mice with day 14 Ad-infected mice, and results were at 3, 7, 14, and 21 days postinfection. Recombinant Ad infec-
then compared between normal and transgenic mouse groups. tion was monitored from GFP marker expression. Hepatocytes
(⬎95%) displayed GFP expression from the recombinant Ad
in NS5A-Tg and NTg mouse livers at day 3 (Fig. 1A). GFP-
RESULTS
positive cells were also readily detectable at days 7, 14, and 21
Impaired clearance of Ad from HCV NS5A-Tg mouse liver. following Ad infection in NS5A-Tg mouse livers in contrast to
To investigate the effect of HCV NS5A on unrelated virus the NTg controls. Liver extracts were prepared from both
clearance from mouse liver, age- and sex-matched NS5A-Tg NS5A-Tg and NTg mice at days 3, 7, 14, and 21 as described
VOL. 83, 2009 HCV NS5A INHIBITS INTRAHEPATIC IFN-␥ PRODUCTION 8465

Downloaded from http://jvi.asm.org/ on May 24, 2017 by guest


FIG. 2. Liver specimens from day 7 Ad-infected NTg and NS5A-Tg mice were immunostained with CD3⫹ antibody and counterstained with
hematoxylin to determine infiltrating T cells (top panels). Liver sections were also stained with hematoxylin and eosin (H&E) to examine
inflammatory infiltrates and liver histopathology (bottom panels).

previously (28), and GFP expression was quantitated using a Intrahepatic cytokine gene expression during Ad infection in
fluorometer (Fig. 1B). GFP expression was normalized to that HCV NS5A-Tg mice. CD4⫹ T cells are a major player in the
from mouse livers on day 3. GFP expression was also examined induction of protective immune response to HCV infection.
by Western blot analysis (Fig. 1C). Together, our results re- Th17 is a newly defined subset of CD4⫹ T cells. The discovery
vealed that the NS5A-Tg mice fail to clear Ad from liver up to of the Th17 lineage has revealed additional complexity in the
21 days postinfection. fates chosen by helper T cells and has begun to reshape our
We have examined the T-cell infiltration in both NTg and view of how signaling and transcriptional networks generate
NS5A-Tg mice at day 7 following Ad infection. For this, the appropriate and inappropriate immunity (37). We have exam-
presence of CD3⫹ cells was examined in liver sections by ined cytokine-related gene expression profiles using real-time
immunohistochemistry. Slides were immunostained with anti- PCR-based focused microarrays for intrahepatic inflammatory
CD3 antibody and counterstained with hematoxylin. CD3⫹ genes related to the Th17 regulatory network. A comparison of
cells from at least five fields of each slide were counted in a intrahepatic cytokines between NS5A-Tg and NTg mice at day
blinded manner. We did not observe a significant difference in 14 of Ad infection is shown in Fig. 3A. Out of 84 cytokine-
the level of CD3 marker between the NS5A-Tg and NTg mice related genes examined, 63 (75.0%) were upregulated by 2.0-
(Fig. 2). The slides were also stained with hematoxylin and
eosin to examine inflammation. We have observed a slightly
higher level of inflammation in NTg mice than in HCV
NS5A-Tg mice following Ad infection. A representative illus- TABLE 1. Serum ALT level in NTg and NS5A-Tg mice
tration is shown in Fig. 2 (bottom panel). Our results are in Time point ALT level (U/liter)a
agreement with the earlier study of transgenic mice expressing (day) NTg mice NS5A-Tg mice
the entire HCV genome (8), suggesting that the presence of
the HCV transgene does not impair intrahepatic T-cell infil- 0 120 ⫾ 31 122 ⫾ 23
7 696 ⫾ 218 188 ⫾ 65
tration. We also examined the alanine aminotransferase 14 1071 ⫾ 206 298 ⫾ 104
(ALT) level from serum of NS5A-Tg and NTg mice at different 24 ND 144 ⫾ 56
time points. The ALT level was increased approximately five- a
Data for each time point represent the sera of six animals, and values are
to eightfold on day 7 and day 14 in NTg mice compared to means ⫾ standard deviations. ND, not done (AdGFP was cleared from mouse
levels in NS5A-Tg mice (Table 1). liver).
8466 KANDA ET AL. J. VIROL.

Downloaded from http://jvi.asm.org/ on May 24, 2017 by guest


FIG. 4. Differential expression of intrahepatic cytokine mRNA lev-
FIG. 3. Differential expression of intrahepatic cytokine genes in els (A) and presence of lymphoid cell markers (B) in Ad-infected the
NS5A-Tg and NTg mice following Ad infection. (A) NTg and liver of NTg and NS5A-Tg mice. Gene expression was measured by
NS5A-Tg mice were infected with Ad or left untreated. Liver RNA quantitative real-time PCR, and results are presented as induction
was isolated at day 14 postinfection and used in focused microarray relative to basal levels in each of the uninfected mice. Six mice in each
analysis for 84 genes. All gene expression levels were corrected by group were examined.
comparison with the level of two housekeeping genes (glyceralde-
hyde-3-phosphate dehydrogenase and actin) and compared. Six sets
of real-time PCR arrays were performed, and the results were both NS5A-Tg and NTg mice infected with Ad (Fig. 4A).
analyzed using RT2 profiler PCR array data analysis software (Su-
perarray). (B) The details of genes upregulated 2.0-fold or greater Expression of chemokine genes, CCL7 (MCP-3) and CX3CR1,
in NTg mice and HCV NS5A-Tg mice are shown. was upregulated to approximately 2.0-fold in NS5A-Tg mice.
Importantly, CCL7 has been implicated with portal inflamma-
tion in liver disease (49) while CX3CR1 is involved in liver
fold or greater in NTg mice while 38 (45%) genes were up- fibrosis due to chronic HCV infection (51).
regulated 2.0-fold or higher in NS5A-Tg mice (Fig. 3B). The Ad-infected NS5A-Tg mice display altered expression of
response to Ad infection was partly related to NS5A expres- intrahepatic lymphoid cell markers. One of the earliest mani-
sion, with 32 genes unique to the NTg mice, 7 unique to festations of a cell-mediated immune response in the infected
NS5A-Tg mice, and 31 expressed in both NS5A-Tg and NTg liver is an increase in lymphoid cells. To examine cell infiltra-
mice. Eleven genes (TNF-␣, interleukin-23 receptor [IL-23R], tion, liver from mice injected with Ad was harvested and ana-
CCL20, granulocyte colony-stimulating factor, IL-21, CXCL5, lyzed for lymphoid cell surface molecules using real-time PCR
IL-12RB2, IL-10, IFN-␥, Toll-like receptor 4 [TLR4], and IL- (Fig. 4B.). Among the T-cell markers, the greatest suppression
17␣) were upregulated fivefold or more in NTg mice, and only was seen in CD8␣ (synonymous for CD8 in human) mRNA,
TNF-␣ was upregulated in NS5A-Tg mice following Ad infec- while CD4 mRNA was suppressed to a smaller extent in
tion. The results of the genes upregulated twofold or greater NS5A-Tg animals. Among the B-cell markers, CD40lg was
are summarized in Fig. 3. On the other hand, out of 84 genes, suppressed in NS5A-Tg animals (Fig. 4B). ICAM-1, a well-
5 (5.9%) genes were downregulated 2.0-fold or more in both known cell adhesion molecule, was upregulated in NS5A-Tg
groups of mice (Fig. 3A). Among these, four genes were mice. Expression of Gata-3, Stat6, Socs3, Syk, Socs1, Jak1,
unique to the NTg mice (IL-17, PBST-1, PB1, and calcyclin Myd88, TLR4, Stat4 and T-bet were upregulated ⱖ2.0-fold in
binding protein) and NS5A-Tg mice (IL-23, CD127, NF-E1, NS5A-Tg mouse livers. Therefore, these results suggested that
and Stat3), and one gene was expressed in both types of mice. NS5A-Tg mouse liver exhibits decreased expression of CD4
CCAAT/enhancer binding protein beta, which has antiapop- and CD8 cell surface molecules. This could be the result of the
totic function, was the only gene downregulated in both animal impact of NS5A expression on the overall ratio of CD4 to CD8
groups. cells and awaits further in-depth analysis. An inverse expres-
Further analysis revealed that among the Th1 cytokines, sion of IFN-␥ and TLR in NS5A-Tg mouse liver is in agree-
IFN-␥, granulocyte colony-stimulating factor, and IL-2 mRNA ment with chronically infected HCV individuals who failed to
expression levels were suppressed significantly in NS5A-Tg respond to IFN therapy (14). Tbx21 modulates IFN-␥ expres-
mice compared to NTg control mice (Fig. 4A). On the other sion in Th1 cells (45). Our results suggested that Tbx21 mRNA
hand, TNF-␣ and IL-12␤ mRNA inductions were similar in expression is suppressed in Ad-infected NS5A-Tg mice. This
VOL. 83, 2009 HCV NS5A INHIBITS INTRAHEPATIC IFN-␥ PRODUCTION 8467

FIG. 5. Ad infection in NS5A-Tg mice does not enhance IFN-␥


secretion. IFN-␥ and TNF-␣ levels were determined by ELISA using
sera from NTg and NS5A-Tg mice. *, P ⬍ 0.001 by the Student’s t test
from comparison between sera of NTg and NS5A-Tg mice.

Downloaded from http://jvi.asm.org/ on May 24, 2017 by guest


result is in agreement with the previous observation that the
Tbx21 mRNA level is reduced in HCV-infected T cells (19). FIG. 6. A schematic diagram depicting the highlights of observa-
Ad infection of NS5A-Tg mice does not enhance IFN-␥ se- tions following Ad challenge in NTg and NS5A-Tg mice.
cretion. TNF-␣ determines the magnitude and course of ac-
quired immune responses. TNF-␣-induced apoptosis is impor-
tant for lysis of virus-infected cells, and the activity of killing TNF-␣ expression is observed in the liver of chronically HCV-
virus-infected cells is accelerated by IFN-␥ (17, 32, 53). Mod- infected individuals (22). Together, our results suggested that
ulation of TNF-␣ and IFN-␥ has been observed in HCV-in- inhibition of IFN-␥ expression in NS5A-Tg mice and blockade
fected patients (16). Since IFN-␥ and TNF-␣ have also been of TNF-␣-mediated apoptosis by NS5A contribute to delayed
implicated in clearance of Ad from mouse liver (17, 43), the Ad clearance. IFN-␥ is not expressed in hepatocytes, and we
modulation of these two cytokines involved in innate signaling therefore believe that a secondary event may modulate IFN-␥
of NS5A-Tg mice was verified at the protein level by ELISA. production since other resident cells cross talk in the liver.
Our results demonstrated that TNF-␣ expression was upregu- IFN-␥ with potential antifibrotic action is associated with the
lated in both NTg and NS5A-Tg mice (Fig. 5). On the other blockage of stellate cell activation, proliferation, and synthesis
hand, IFN-␥ production was significantly lower in Ad-infected of collagen. Experimental studies have demonstrated that
NS5A-Tg mice. These results are in agreement with our quan- IFN-␥ inhibits extracellular matrix mRNA expression (47).
titative reverse transcription-PCR data described above and Suppression of IFN-␥ by NS5A may also be associated with the
the observations from chronically HCV-infected humans (35). progression of hepatic fibrosis in chronically infected HCV
patients. Our data support the earlier observation that patients
DISCUSSION with self-limited hepatitis C display the strongest virus-specific
CD4⫹ T-cell (IFN-␥) reactivity while persistently infected sub-
We have examined whether HCV NS5A interferes with the jects initially show a weak antiviral CD4⫹ T-cell (IFN-␥) re-
innate antiviral signaling pathway in liver. For this, unrelated sponse (14). We did not observe significant poly(ADP-ribose)
hepatotropic Ad clearance from NS5A-Tg mouse liver was polymerase cleavage in Ad-infected NS5A-Tg mouse liver ex-
investigated. The Ad-dependent antiviral effect occurs in two tract, suggesting an absence of apoptosis.
distinct phases and has been eloquently described by Ca- Gata-3 is essential for NK cell homing to the liver. While
vanaugh et al. (5). The first phase of induction occurs within 12 Gata-3 antagonizes IFN-␥ production in differentiating T cells,
to 24 h of infection and is mediated by IFN-␣/␤. The second Gata-3-deficient NK cells paradoxically produce less IFN-␥
phase occurs after 3 to 7 days of infection and is associated than control NK cells (37, 38). Gata-3 serves as a principal
with intrahepatic induction of IFN-␣/␤ and TNF-␣ as well as switch in determining Th1/Th2 responses (54). Optimal induc-
IFN-␥ for induction of the cellular immune response to the tion of IFN-␥ expression also requires genetic interaction be-
invading virus. The production of these cytokines was also seen tween Tbx21 (T-bet) and its target, the homeoprotein Hlx (34).
in human infection from Ad and plays an important role in Tbx21 expression correlates with IFN-␥ expression in Th1 and
viral clearance (30, 33, 43). An earlier report suggested that NK cells (45). Tbx21 mRNA was 2.2-fold upregulated in
transgenic mice expressing the entire HCV genome delay Ad NS5A-Tg mice and 8.7-fold upregulated in NTg mice following
clearance from liver (8). On the other hand, mice expressing Ad infection. These results are in agreement with the obser-
HCV structural proteins do not impair Ad clearance (24, 42). vation that the Tbx21 mRNA level is suppressed in HCV-
Here, we have shown that NS5A-Tg mice modulate the innate infected T cells (19). Tbx21 is required for protection against
immune response in the liver and impair Ad clearance. Inter- human viral infection (6, 21, 31, 44). The principal function of
estingly, both NTg and NS5A-Tg mice displayed elevation of Tbx21 in developing Th1 cells is to negatively regulate Gata-3
TNF-␣ while the level of IFN-␥ was significantly lower in (50). T lymphocytes have an essential role in the clearance of
NS5A-Tg mice following Ad infection. However, HCV NS5A acute HCV infection (7, 23). Individuals who are able to suc-
blocks TNF-␣-mediated apoptosis (29), and a high level of cessfully clear HCV seem to have not only specific but also
8468 KANDA ET AL. J. VIROL.

broad cellular Th1-dominant responses. Abundant evidence ential expression of toll-like receptor mRNA in treatment non-responders
and sustained virologic responders at baseline in patients with chronic hep-
indicates that the immune system fails to eliminate virus from atitis C. Liver Int. 26:1100–1110.
the liver in a large number of HCV-infected patients, and as a 16. Huang, Y., J. J. Feld, R. K. Sapp, S. Nanda, J. H. Lin, L. M. Blatt, M. W.
result, most acute infections lead to long-term viral persistence Fried, K. Murthy, and T. J. Liang. 2007. Defective hepatic response to
interferon and activation of suppressor of cytokine signaling 3 in chronic
(12, 36). The efficacies of therapeutic vaccination for chronic hepatitis C. Gastroenterology 132:733–744.
hepatitis C nonresponder patients are correlated with the in- 17. Kafrouni, M., G. R. Brown, and D. L. Thiele. 2003. The role of TNF-TNFR2
hibition of IFN-␥ secretion (3, 18, 34). Our results revealed interactions in generation of CTL responses and clearance of hepatic ade-
novirus infection. J. Leukoc. Biol. 74:564–571.
that HCV NS5A affects Gata-3 and Tbx21 expression and 18. Klade, C. S., H. Wedemeyer, T. Berg, H. Hinrichsen, G. Cholewinska, S.
impairs IFN-␥ production (Fig. 6), thus contributing to viral Zeuzem, H. Blum, M. Buschle, S. Jelovcan, V. Buerger, E. Tauber, J. Frisch,
and M. P. Manns. 2008. Therapeutic vaccination of chronic hepatitis C
persistence. nonresponder patients with the peptide vaccine IC41. Gastroenterology 134:
1385–1395.
ACKNOWLEDGMENTS 19. Kondo, Y., V. M. Sung, K. Machida, M. Liu, and M. M. Lai. 2007. Hepatitis
C virus infects T cells and affects interferon-gamma signaling in T cell lines.
We thank Brendan Kappas for PCR arrays and John E. Sagartz for Virology 361:161–173.
helping us to evaluate the immunohistochemistry slides. 20. Koziel, M. J. 1999. Cytokines in viral hepatitis. Semin. Liver Dis. 19:157–169.
This work was supported by research grants AI45144 and AI065535 21. Kulkarni, A., D. S. Ravi, K. Singh, S. Rampalli, V. Parekh, D. Mitra, and S.
(R.B.R.) from the National Institutes of Health and by the Blue Rib- Chattopadhyay. 2005. HIV Tat modulates T-bet expression and induces Th1
type of immune response. Biochem. Biophys. Res. Commun. 329:706–712.

Downloaded from http://jvi.asm.org/ on May 24, 2017 by guest


bon Fund from Saint Louis University. 22. Larrea, E., N. Garcia, C. Qian, M. P. Civeira, and J. Prieto. 1996. Tumor
necrosis factor alpha gene expression and the response to interferon in
REFERENCES chronic hepatitis C. Hepatology 23:210–217.
1. Abe, T., Y. Kaname, I. Hamamoto, Y. Tsuda, X. Wen, S. Taguwa, K. Mori- 23. Lechner, F., D. K. Wong, P. R. Dunbar, R. Chapman, R. T. Chung, P.
ishi, O. Takeuchi, T. Kawai, T. Kanto, N. Hayashi, S. Akira, and Y. Mat- Dohrenwend, G. Robbins, R. Phillips, P. Klenerman, and B. D. Walker.
suura. 2007. Hepatitis C virus nonstructural protein 5A modulates the Toll- 2000. Analysis of successful immune responses in persons infected with
like receptor–MyD88-dependent signaling pathway in macrophage cell lines. hepatitis C virus. J. Exp. Med. 191:1499–1512.
J. Virol. 81:8953–8966. 24. Liu, Z.-X., H. Nishida, J.-W. He, M. M. C. Lai, N. Feng, and G. Dennert.
2. Abougergi, M. S., S. J. Gidner, D. K. Spady, B. C. Miller, and D. L. Thiele. 2002. Hepatitis C virus genotype 1b core protein does not exert immuno-
2005. Fas and TNFR1, but not cytolytic granule-dependent mechanisms, modulatory effects on virus-induced cellular immunity. J. Virol. 76:990–997.
mediate clearance of murine liver adenoviral infection. Hepatology 41:97– 25. MacQuillan, G. C., C. Mamotte, W. D. Reed, G. P. Jeffrey, and J. E. Allan.
105. 2003. Upregulation of endogenous intrahepatic interaction stimulated genes
3. Alvarez-Lajonchere, L., N. H. Shoukry, B. Gra, Y. Amador-Canizares, F. during chronic hepatitis C virus infection. J. Med. Virol. 70:219–227.
Helle, N. Bedard, I. Guerra, C. Drouin, J. Dubuisson, E. E. Gonzalez-Horta, 26. Major, M. E., H. Dahari, K. Mihalik, M. Puig, C. M. Rice, A. U. Neumann,
G. Martinez, J. Marante, Z. Cinza, M. Castellanos, and S. Duenas-Carrera. and S. M. Feinstone. 2004. Hepatitis C virus kinetics and host responses
2009. Immunogenicity of CIGB-230, a therapeutic DNA vaccine prepara- associated with diseases and outcome of infection in chimpanzees. Hepatol-
tion, in HCV-chronically infected individuals in a Phase I clinical trial. J. ogy 39:1709–1720.
Viral Hepat. 16:156–167. 27. Majumder, M., A. K. Ghosh, R. Steele, R. Ray, and R. B. Ray. 2001. Hep-
4. Bigger, C. B., B. Guerra, K. M. Brasky, G. Hubbard, M. R. Beard, B. A. atitis C virus NS5A physically associates with p53 and regulates p21/waf1
Luxon, S. M. Lemon, and R. E. Lanford. 2004. Intrahepatic gene expression gene expression in a p53-dependent manner. J. Virol. 75:1401–1407.
during chronic hepatitis C virus infection in chimpanzees. J. Virol. 78:13779– 28. Majumder, M., A. K. Ghosh, R. Steele, X. Y. Zhou, N. J. Phillips, R. Ray, and
13792. R. B. Ray. 2002. Hepatitis C virus NS5A protein impairs TNF-mediated
5. Cavanaugh, V. J., L. G. Guidotti, and F. V. Chisari. 1998. Inhibition of hepatic apoptosis, but not by an anti-FAS antibody, in transgenic mice.
hepatitis B virus replication during adenovirus and cytomegalovirus infec- Virology 294:94–105.
tions in transgenic mice. J. Virol. 72:2630–2637. 29. Majumder, M., R. Steele, A. K. Ghosh, X. Y. Zhou, L. Thornburg, R. Ray,
6. Chen, R. F., J. W. Liu, W. T. Yeh, L. Wang, J. C. Chang, H. R. Yu, J. T. N. J. Phillips, and R. B. Ray. 2003. Expression of hepatitis C virus non-
Cheng, and K. D. Yang. 2005. Altered T helper 1 reaction but not increase structural 5A protein in the liver of transgenic mice. FEBS Lett. 555:528–
of virus load in patients with dengue hemorrhagic fever. FEMS Immunol. 532.
Med. Microbiol. 44:43–50. 30. Matsubara, T., T. Inoue, N. Tashiro, K. Katayama, T. Matsuoka, and S.
7. Day, C. L., G. M. Lauer, G. K. Robbins, B. McGovern, A. G. Wurcel, R. T. Furukawa. 2000. Activation of peripheral blood CD8⫹ T cells in adenovirus
Gandhi, R. T. Chung, and B. D. Walker. 2002. Broad specificity of virus infection. Pediatr. Infect. Dis. J. 19:766–768.
specific CD4⫹ T-helper-cell responses in resolved hepatitis C virus infection. 31. Matsui, M., O. Moriya, T. Yoshimoto, and T. Akatsuka. 2005. T-bet is
J. Virol. 76:12584–12595. required for protection against vaccinia virus infection. J. Virol. 79:12798–
8. Disson, O., D. Haouzi, S. Desagher, K. Loesch, M. Hahne, E. J. Kremer, C. 12806.
Jacquet, S. M. Lemon, U. Hibner, and H. Lerat. 2004. Impaired clearance of 32. Mestan, J., W. Digel, S. Mittnacht, H. Hillen, D. Blohm, A. Moller, H.
virus-infected hepatocytes in transgenic mice expressing the hepatitis C virus Jacobsen, and H. Kirchner. 1986. Antiviral effects of recombinant tumor
polyprotein. Gastroenterology 126:859–872. necrosis factor in vitro. Nature 323:816–819.
9. Frese, M., K. Barth, A. Kaul, V. Lohmann, V. Schwarzle, and R. Barten- 33. Mistchenko, A. S., R. A. Diez, A. L. Mariani, J. Robaldo, A. F. Maffey, G.
schlager. 2002. Interferon-gamma inhibits replication of subgenomic and Bayley-Bustamante, and S. Grinstein. 1994. Cytokines in adenoviral disease
genomic hepatitis C virus RNAs. Hepatology 35:694–703. in children: association of interleukin-6, interleukin-8, and tumor necrosis
10. Ghosh, A. K., I. Grigorieva, R. Steele, R. G. Hoover, and R. B. Ray. 1999. factor alpha levels with clinical outcome. J. Pediatr. 124:714–720.
PTEN transcriptionally modulates c-myc gene expression in human breast 34. Mullen, A. C., A. S. Hutchins, F. A. High, H. W. Lee, K. J. Sykes, L. A.
carcinoma cells and is involved in cell growth regulation. Gene 235:85–91. Chodosh, and S. L. Reiner. 2002. Hlx is induced by and genetically interacts
11. Ghosh, A. K., M. Majumder, R. Steele, K. Meyer, R. Ray, and R. B. Ray. with T-bet to promote heritable TH1 gene induction. Nat. Immunol. 3:652–
2000. Hepatitis C virus NS5A protein protects against TNF-alpha mediated 658.
apoptotic cell death. Virus Res. 67:173–178. 35. Neveu, B., E. Debeaupuis, K. Echasserieau, B. le Moullac-Vaidye, M. Gas-
12. Graham, C. S., M. Curry, Q. He, N. Afdhal, D. Nunes, C. Fleming, R. sin, L. Jegou, J. Decalf, M. Albert, N. Ferry, J. Gournay, E. Houssaint, M.
Horsburgh, D. Craven, K. E. Sherman, and M. J. Koziel. 2004. Comparison Bonneville, and X. Saulquin. 2008. Selection of high-avidity CD8 T cells
of HCV-specific intrahepatic CD4⫹ T cells in HIV/HCV versus HCV. Hepa- correlates with control of hepatitis C virus infection. Hepatology 48:713–722.
tology 40:125–132. 36. Rahman, F., T. Heller, Y. Sobao, E. Mizukoshi, M. Nascimbeni, H. Alter, S.
13. Gruner, N. H., T. J. Gerlach, M.-C. Jung, H. M. Diepolder, C. M. Schirren, Herrine, J. Hoofnagle, T. J. Liang, and B. Rehermann. 2004. Effects of
W. W. Schraut, R. Hoffmann, R. Zachoval, T. Santantonio, M. Cucchiarini, antiviral therapy on the cellular immune response in acute hepatitis C.
A. Cerny, and G. R. Pape. 2000. Association of hepatitis C virus-specific Hepatology 40:87–97.
CD8⫹ T cells with viral clearance in acute hepatitis C. J. Infect. Dis. 181: 37. Reiner, S. L. 2007. Development in motion: helper T cells at work. Cell
1528–1536. 129:33–36.
14. Guobuzaite, A., S. Chokshi, L. Balciūniene, A. Voinic, A. Stikleryte, K. 38. Samson, S. I., O. Richard, M. Tavian, T. Ranson, C. A. Vosshenrich, F.
Zagminas, A. Ambrozaitis, and N. Naoumov. 2008. Viral clearance or per- Colucci, F. J. Buer, F. Grosveld, I. Godin, and J. P. Di Santo. 2003. GATA-3
sistence after acute hepatitis C infection: interim results from a prospective promotes maturation, IFN-gamma production, and liver-specific homing of
study. Medicina (Kaunas) 44:510–520. NK cells. Immunity 19:701–711.
15. He, Q., C. S. Graham, E. Durante Mangoni, and M. J. Koziel. 2006. Differ- 39. Sarcar, B., A. K. Ghosh, R. Steele, R. Ray, and R. B. Ray. 2004. Hepatitis C
VOL. 83, 2009 HCV NS5A INHIBITS INTRAHEPATIC IFN-␥ PRODUCTION 8469

virus NS5A mediated STAT3 activation requires co-operation of Jak1 ki- of hepatitis C virus infection and hepatic fibrosis: new insight into antifibrotic
nase. Virology 322:51–60. therapy in chronic hepatitis C. Hepatol. Res. 37:579–595.
40. Schmittgen, T. D., B. A. Zakrajsek, A. G. Mills, V. Gorn, M. J. Singer, and 48. Thimme, R., J. Bukh, H. C. Spangenberg, S. Wieland, J. Pemberton, C.
M. W. Reed. 2000. Quantitative reverse transcription-polymerase chain re- Steiger, S. Govindarajan, R. H. Purcell, and F. V. Chisari. 2002. Viral and
action to study mRNA decay: comparison of endpoint and real-time meth- immunological determinants of hepatitis C virus clearance, persistence, and
ods. Anal. Biochem. 285:194–204. disease. Proc. Natl. Acad. Sci. USA 99:15661–15668.
41. Su, A. I., J. P. Pezacki, L. Wodicka, A. D. Brideau, L. Supekova, R. Thimme, 49. Tsuneyama, K., K. Harada, M. Yasoshima, K. Hiramatsu, C. R. Mackay,
S. Wieland, J. Bukh, R. H. Purcell, P. G. Schultz, and F. V. Chisari. 2002. I. R. Mackay, M. E. Gershwin, and Y. Nakanuma. 2001. Monocyte chemo-
Genomic analysis of the host response to hepatitis C virus infection. Proc. tactic protein-1, -2, and -3 are distinctively expressed in portal tracts and
Natl. Acad. Sci. USA 99:15669–15674. granulomata in primary biliary cirrhosis: implications for pathogenesis.
42. Sun, J., F. Bodola, X. Fan, H. Irshad, L. Soong, S. M. Lemon, and T. S. J. Pathol. 193:102–109.
Chan. 2001. Hepatitis C virus core and envelope proteins do not suppress the 50. Usui, T., J. C. Preiss, Y. Kanno, Z. J. Yao, J. H. Bream, J. J. O’Shea, and W.
host’s ability to clear a hepatic viral infection. J. Virol. 75:11992–11998. Strober. 2006. T-bet regulates Th1 responses through essential effects on
GATA-3 function rather than on IFNG gene acetylation and transcription.
43. Sung, R. S., L. Qin, and J. S. Bromberg. 2001. TNF␣ and IFN␥ induced by
J. Exp. Med. 203:755–766.
innate anti-adenoviral immune responses inhibit adenovirus-mediated trans-
51. Wasmuth, H. E., M. M. Zaldivar, M. L. Berres, A. Werth, D. Scholten, S.
gene expression. Mol. Ther. 3:757–767.
Hillebrandt, F. Tacke, P. Schmitz, E. Dahl, T. Wiederholt, C. Hellerbrand,
44. Svensson, A., I. Nordstrom, J. B. Sun, and K. Eriksson. 2005. Protective T. Berg, R. Weiskirchen, C. Trautwein, and F. Lammert. 2008. The frac-
immunity to genital herpes simplex virus type 2 infection is mediated by talkine receptor CX3CR1 is involved in liver fibrosis due to chronic hepatitis
T-bet. J. Immunol. 174:6266–6273. C infection. J. Hepatol. 48:208–215.
45. Szabo, S. J., S. T. Kim, G. L. Costa, X. Zhang, C. G. Fathman, and L. H. 52. Wieland, S. F., and F. V. Chisari. 2005. Stealth and cunning: hepatitis B and
Glimcher. 2000. A novel transcription factor, T-bet, directs Th1 lineage hepatitis C viruses. J. Virol. 79:9369–9380.

Downloaded from http://jvi.asm.org/ on May 24, 2017 by guest


commitment. Cell 100:655–669. 53. Wong, G. H., and D. V. Goeddel. 1986. Tumor necrosis factors alpha and beta
46. Tanji, Y., T. Kaneko, S. Satoh, and K. Shimotohno. 1995. Phosphorylation of inhibit virus replication and synergize with interferons. Nature 323:819–822.
hepatitis C virus-encoded nonstructural protein NS5A. J. Virol. 69:3980– 54. Zhu, J., B. Min, C. J. Watson, A. Grinberg, Q. Wang, N. Killeen, J. F. Urban
3986. Jr., L. Guo, and W. E. Paul. 2004. Conditional deletion of Gata-3 shows its
47. Teixeira, R., L. A. Marcos, and S. L. Friedman. 2007. Immunopathogenesis essential function in TH1-TH2 responses. Nat. Immunol. 5:1157–1165.

You might also like