ArticlePDF Available

Hepatic Gene Expression During Treatment with Peginterferon and Ribavirin: Identifying Molecular Pathways for Treatment Response

Authors:

Abstract and Figures

Unlabelled: The reasons for hepatitis C treatment failure remain unknown but may be related to different host responses to therapy. In this study, we compared hepatic gene expression in patients prior to and during peginterferon and ribavirin therapy. In the on-treatment group, patients received either ribavirin for 72 hours prior to peginterferon alpha-2a injection or peginterferon alpha-2a for 24 hours, prior to biopsy. The patients were grouped into rapid responders (RRs) with a greater than 2-log drop and slow responders (SRs) with a less than 2-log drop in hepatitis C virus RNA by week 4. Pretreatment biopsy specimens were obtained from a matched control group. The pretreatment patients were grouped as RRs or SRs on the basis of the subsequent treatment response. Gene expression profiling was performed with Affymetrix microarray technology. Known interferon-stimulated genes (ISGs) were induced in treated patients. In the pretreatment group, future SRs had higher pretreatment ISG expression than RRs. On treatment, RRs and SRs had similar absolute ISG expression, but when it was corrected for the baseline expression with the pretreatment group, RRs showed a greater fold change in ISGs, whereas SRs showed a greater change in interferon (IFN)-inhibitory pathways. The patients pretreated with ribavirin had heightened induction of IFN-related genes and down-regulation of genes involved in IFN inhibition and hepatic stellate cell activation. Conclusion: These data suggest that ISG inducibility is important for the treatment response and that ribavirin may improve outcomes by enhancing hepatic gene responses to peginterferon. Collectively, these mechanisms may provide a molecular basis for the improved efficacy of combination therapy.
Content may be subject to copyright.
Hepatic Gene Expression During Treatment with Peginterferon
and Ribavirin: Identifying Molecular Pathways for Treatment
Response
Jordan J. Feld1, Santosh Nanda1, Ying Huang1, Weiping Chen2, Maggie Cam2, Susan N.
Pusek3, Lisa M. Schweigler3, Dickens Theodore3, Steven L. Zacks3, T. Jake Liang1,*, and
Michael W. Fried3,*
1 Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National
Institutes of Health, Bethesda, MD
2 Microarray Facility, National Institute of Diabetes and Digestive and Kidney Diseases, National
Institutes of Health, Bethesda, MD
3 University of North Carolina, Chapel Hill, NC
Abstract
The reasons for hepatitis C treatment failure remain unknown but may be related to different host
responses to therapy. In this study, we compared hepatic gene expression in patients prior to and
during peginterferon and ribavirin therapy. In the on-treatment group, patients received either
ribavirin for 72 hours prior to peginterferon alpha-2a injection or peginterferon alpha-2a for 24 hours,
prior to biopsy. The patients were grouped into rapid responders (RRs) with a greater than 2-log drop
and slow responders (SRs) with a less than 2-log drop in hepatitis C virus RNA by week 4.
Pretreatment biopsy specimens were obtained from a matched control group. The pretreatment
patients were grouped as RRs or SRs on the basis of the subsequent treatment response. Gene
expression profiling was performed with Affymetrix microarray technology. Known interferon-
stimulated genes (ISGs) were induced in treated patients. In the pretreatment group, future SRs had
higher pretreatment ISG expression than RRs. On treatment, RRs and SRs had similar absolute ISG
expression, but when it was corrected for the baseline expression with the pretreatment group, RRs
showed a greater fold change in ISGs, whereas SRs showed a greater change in interferon (IFN)-
inhibitory pathways. The patients pretreated with ribavirin had heightened induction of IFN-related
genes and down-regulation of genes involved in IFN inhibition and hepatic stellate cell activation.
Conclusion—These data suggest that ISG inducibility is important for the treatment response and
that ribavirin may improve outcomes by enhancing hepatic gene responses to peginterferon.
Collectively, these mechanisms may provide a molecular basis for the improved efficacy of
combination therapy.
Despite great advances in the treatment of chronic hepatitis C infection, the current therapy
with peginterferon and ribavirin is effective in only about 50% of patients.1,2 The reasons for
treatment failure are not well understood but likely are related to both viral and host factors.3
Address reprint requests to: T. Jake Liang, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases,
National Institutes of Health, Building 10, Room 9B16, 10 Center Drive, MSC 1800, Bethesda MD 20892-1800.
jakel@intra.niddk.nih.gov; fax: 301 402 0491.
*These authors contributed equally to this study.
Potential conflict of interest: Dr. Fried is a consultant for and received grants from Roche. Dr. Zacks is on the speakers’ bureau of Roche.
Supplementary material for this article can be found on the Hepatology Web site
(http://interscience.wiley.com/jpages/0270-9139/suppmat/index.html).
NIH Public Access
Author Manuscript
Hepatology. Author manuscript; available in PMC 2010 January 19.
Published in final edited form as:
Hepatology. 2007 November ; 46(5): 1548–1563. doi:10.1002/hep.21853.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
The viral genotype has the greatest impact on the treatment outcome. A sustained virological
response (SVR) is achieved in 42%-46% of genotype 1 infections after a year of therapy, in
contrast to rates of 80% in genotype 2 and 3 infections after just 6 months of treatment.1,2 In
addition to the genotype, the baseline viral load is also an important factor, particularly in
genotype 1 infection.2 Although numerous viral strategies for interfering with host viral
defense mechanisms have been identified, none has been clearly shown to be responsible for
the genotypic differences in the treatment response.
From large treatment trials, a number of host factors have been found to be associated with the
treatment response. Gender and race are the most important factors. Men consistently respond
less well to therapy than women, and African Americans have poorer outcomes than Caucasian
populations.4 Age, obesity, and the degree of liver fibrosis also affect the treatment outcome.
5 Although these factors have been consistently identified in multiple studies, the mechanism
by which they affect the treatment outcome remains unknown.
To gain a further understanding of the host factors, microarray technology has been used to
evaluate hepatic gene expression prior to antiviral therapy. Chen et al.6 found that pretreatment
gene expression profiles from liver biopsies were predictive of the ultimate treatment outcome.
Patients who did not respond to therapy showed up-regulation of numerous interferon-
stimulated genes (ISGs) prior to treatment in comparison with both sustained responders and
normal controls. Hepatic gene expression has not been reported in humans undergoing therapy.
Several studies have reported gene expression in peripheral blood mononuclear cells (PBMCs)
during the course of therapy; however, gene induction in PBMCs may not be entirely reflective
of events in the liver.79
The addition of ribavirin to interferon (IFN) therapy significantly improves the treatment
response rates; however, the mechanism by which this occurs is poorly understood. Numerous
mechanisms of action for ribavirin have been proposed, including inosine-5-monophosphate
dehydrogenase inhibition (IMPDH), direct viral inhibition, increased mutagenesis leading to
error catastrophe, and promotion of a Th1 immune response.10 Although there is some
experimental evidence to support all of these mechanisms, none accounts for the magnitude
of the benefit seen with the addition of ribavirin.
In order to gain further understanding of the genetic factors that may contribute to the treatment
response, we evaluated gene expression from liver biopsy samples from patients currently
undergoing treatment with peginterferon. Half the patients also received ribavirin prior to liver
biopsy, and this allowed us to assess the contribution of this agent to gene expression.
Expression profiles from on-treatment patients were compared with those from pretreatment
liver biopsy samples of a matched control population.
Materials and Methods
Study Subjects
Adult patients evaluated at the University of North Carolina Liver Clinic and infected with
hepatitis C virus (HCV) genotype 1 were eligible for enrollment. Patients with a hepatitis B or
human immunodeficiency virus coinfection or a major systemic illness were excluded. All
patients received 180 μg of peginterferon alpha-2a subcutaneously 24 hours prior to liver
biopsy. The patients were randomized to receive peginterferon alone or to receive ribavirin
prior to biopsy as well. Those randomized to receive ribavirin were given weight-based
ribavirin (1000 mg/day for patients weighing less than 75 kg and 1200 mg/day for patients
weighing more than 75 kg) for 72 hours prior to liver biopsy. A sample of biopsy tissue was
snap-frozen in liquid nitrogen and stored at 80°C. The remainder of the biopsy was placed in
formalin for a histological evaluation. After the liver biopsy, the patients continued on 180
Feld et al. Page 2
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
μg of peginterferon per week plus weight-based ribavirin for 48 weeks (Fig. 1). All patients
agreed to undergo a liver biopsy after starting antiviral therapy and signed an informed consent
form. This investigator-initiated study was approved by the Institutional Review Board at the
University of North Carolina Medical Center.
The HCV viral load was measured with Roche Taq-Man (Roche Molecular Systems,
Alamaeda, CA) prior to therapy and serially during therapy. Those patients achieving at least
a 2-log drop in HCV RNA by 4 weeks of therapy were deemed rapid responders (RRs). Patients
with less than a 2-log drop in HCV RNA at 4 weeks were deemed slow responders (SRs).
Standard definitions for relapse (HCV RNA–negative at 48 weeks with subsequent recurrent
viremia), nonresponse [persistently HCV RNA–positive by polymerase chain reaction (PCR)
throughout therapy], and SVR (HCV RNA–negative 6 months after the completion of therapy)
were used to define the ultimate treatment outcome.
Stored liver tissue from pretreatment liver biopsies of patients with genotype 1 HCV infection
performed at the Clinical Center of the National Institutes of Health were used as pretreatment
controls. The same exclusion criteria were used for the pretreatment population, and patients
were selected to match on-treatment patients for gender, race, age, ultimate treatment outcome,
HCV genotype, baseline HCV viral load, and liver biopsy histological scores. Biopsy samples
were snap-frozen in liquid nitrogen at the time of biopsy and stored at 80°C. All but 4 of the
pretreatment patients were subsequently treated, and the on-treatment viral kinetics and
treatment response were known. The patients were categorized as RRs or SRs with the same
definitions used for the on-treatment group (rapid response 2-log drop in HCV RNA within
the first 4 weeks of therapy, slow response < 2-log drop by 4 weeks).
RNA Extraction, Amplification, and Microarray Analysis
Hepatic tissue was placed in Trizol and mechanically ground with a piston until it was
dissolved. RNA was then extracted with the RNeasy kit from Qiagen (Valencia, CA) according
to the manufacturer’s instructions. RNA was quantified with a spectrophotometer, and the RNA
quality was analyzed with an Agilent (Foster City, CA) bioanalyzer according to the
manufacturer’s instructions. RNA was then amplified with an Agilent Enzo kit. Amplified
complementary RNA was hybridized to an Affymetrix Human 133 Plus 2.0 microarray chip
containing 54,675 gene transcripts. The chips were scanned, and the signal intensity was
evaluated as previously described.11
Quantitative Real-Time PCR
A total of 14 genes were selected for real-time PCR confirmation and consisted of genes from
major pathways identified by the microarray analysis. The selected genes were divided into 4
categories: (1) IFN-related genes [IFN-alpha receptor 2, interferon regulatory factor 7 (IRF7),
ISG-15, signal transducer and activator of transcription 1 (STAT1), oligoadenylate synthetase
3 (OAS3), and myxovirus resistance 1 (Mx1)], (2) IFN-inhibitory genes [protein inhibitor of
activated signal transducer and activator of transcription 4 (PIAS4) and protein phosphatase
2a catalytic subunit (PP2Ac)], (3) apoptosis-related genes (Fas and cytochrome C), and (4)
genes related to hepatic stellate cell (HSC) activation [collagen type 1 alpha 2, CD36 (collagen
type 1 receptor), and tissue inhibitor of metallopeptidase 2 (TIMP2)]. Primers and probes were
obtained from Applied Biosystems (Foster City, CA). Real-time PCR was performed with
TaqMan technology as described.12 All values were normalized for the glyceraldehyde 3-
phosphate dehydrogenase expression level. All samples were repeated in duplicate, and mean
expression values were used.
TaqMan confirmation was performed for all patient samples with adequate remaining RNA
after the microarray analysis. In addition, a group of patient samples with inadequate RNA for
Feld et al. Page 3
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the microarray analysis were included for real-time quantitative PCR. Samples analyzed only
by PCR were divided by the treatment group and response similarly to those used for the
microarray analysis. The gene expression patterns identified for confirmation by TaqMan were
based on the initial cohort for which there was sufficient RNA to perform the microarray
analysis. Because the latter group, for which only TaqMan was performed, was not included
in the original microarray analysis, it served as a validation cohort.
Statistical Analysis
The signal intensity from gene transcripts was compared between groups with Partek (St. Louis,
MO) software and Affymetrix (United States) MAS5 normalized log-signal comparisons.
Before the groups were compared, the signal-to-noise ratio was evaluated by a source of
variance analysis. This evaluates whether intergroup variation is greater than intragroup
variation across all comparisons. Further analysis was considered only for comparisons in
which between-group variation was significantly greater than within-group variation. For each
comparison, models were developed to evaluate the effects of race and sex as confounding
variables. The final selected model maximized the intergroup-to-intragroup differences or
signal-to-noise ratio by minimizing the error of the model. For example, when the treatment
response in the pretreatment group was compared, gender, race, and the interaction terms of
gender, race, and response were evaluated in the model. The inclusion of gender and the
interaction of gender and treatment response improved the model, giving an increased signal-
to-noise ratio. The addition of race had no effect on the model and thus was not included in
the final analysis. Similar model development was performed for all comparisons. After the
best model was selected, groups were compared with an analysis of variance. Gene expression
was compared between the treatment groups and by the treatment outcome. Only genes for
which a signal was detected in at least 50% of the samples were included. Expression
differences of at least 1.5-fold with P < 0.01 were considered significant. Because of the
significant risk of false positives with this type of analysis for individual gene expression,
known pathways were also compared with software from GeneGo, Inc. (Michigan). This
evaluates the gene expression of all genes in an established signal transduction or other
molecular pathway. The correction of an analysis of variance with Bonferroni or other
corrections for multiple comparisons generally assumes the complete independence of each
comparison. However, for a microarray analysis, this assumption is unlikely to be correct. If,
for example, 7 genes in a particular pathway are up-regulated or down-regulated, this is much
more likely to be a valid finding than if 7 random genes with unrelated functions are found to
be differentially regulated. Although all genes found by fold-change and P-value cutoffs are
listed, inferences were restricted to genes identified by a pathway analysis to be differentially
regulated. Once gene lists for each comparison were established, supervised hierarchical
clustering and heat maps were created with Partek software.
Results
Sufficient RNA with adequate quality for microarray analysis was available from 11 patients
pretreated with peginterferon with or without ribavirin (on-treatment group) and from 19
patients with biopsies prior to therapy (pretreatment group; Table 1). The groups were matched
by gender, race, and age, and all patients had a genotype 1 infection. The groups were also well
matched for the initial HCV viral load and histological grade and stage (Table 1). Six patients
in the on-treatment group achieved a rapid response (>2-log drop of HCV RNA by 4 weeks),
whereas 5 had a slow response. Six treated patients ultimately achieved an SVR (5 with a rapid
response and 1 with a slow response), 3 relapsed (all with a slow response), and 2 were lost to
follow-up (1 with a rapid response and 1 with a slow response). In the pretreatment group, 5
patients had a rapid response, 10 had a slow response, and 4 were treatment-naive; 5 patients
had an SVR (all with a rapid response), and 10 were nonresponders (all with a slow response).
Feld et al. Page 4
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Six patients in the on-treatment group were treated with ribavirin in addition to peginterferon
prior to liver biopsy, 4 of whom went on to achieve SVR. The additional patients used for real-
time PCR for whom inadequate RNA was available for microarray analysis are described in
Table 1.
On-Treatment versus Pretreatment
Race and gender were found to contribute to the model for this comparison. After we controlled
for the effects of gender and race, 6017 genes were differentially regulated between the on-
treatment and pretreatment groups. Of genes that had detectable microarray signals in greater
than 50% of the samples, a total of 364 genes differed by greater than 1.5-fold expression with
a P value of less than 0.01. A summary of differing gene expression is shown in Table 2 and
highlighted in a heat map in Fig. 2. A full list of genes is available in the supplementary material
(Supplement to Table 2). Known ISGs were induced in the on-treatment group in comparison
with the pretreatment group. Classical ISGs such as OAS3, Mx1, and ISG15 and IRFs were
up-regulated. Other genes with known antiviral activities, including viperin, adenosine
deaminase RNA-specific, phospholipid scramblase, and apolipoprotein B messenger RNA
editing enzyme catalytic polypeptide-3A, were also induced in treated patients.13 ISGs provide
the effector antiviral functions of the IFN response. Genes involved in IFN production,
including IRF7 and retinoic acid–inducible gene I (RIG-I), were induced in the treated patients.
In addition to known ISGs, genes involved in the immune response, including interleukins,
chemokines, major histocompatibility complex class I, and nonspecific factors such as beta-2-
microglobulin and C-reactive protein, were also induced by peginterferon treatment. A
pathway analysis revealed that genes involved in antigen presentation, oxidative stress, and
apoptosis were also significantly up-regulated in on-treatment patients (Fig. 3). Although a
number of genes were also down-regulated by the treatment, aside from genes involved in
cellular proliferation such as the ras-oncogene family and tetraspanins, no clear pattern was
apparent.
Treatment Response
To evaluate whether pretreatment biopsies are of use for predicting treatment response, the
future SRs (same as nonresponders) and RRs (same as sustained responders) in the pretreatment
group were compared. The best model for this comparison included gender but not race. After
we controlled for the effect of gender, 2765 genes were differentially regulated, and 220 were
detectable in greater than 50% of the samples and differed by 1.5-fold or more with a P value
of 0.01 or less. As previously reported,6 nonresponders had significantly higher expression of
numerous ISGs than those who achieved an SVR when treated (Table 3 and Supplement to
Table 3).
Gene expression patterns in patients who received treatment prior to liver biopsy were
compared on the basis of their treatment response. Patients who achieved a 2-log or greater
drop in HCV RNA by 4 weeks of therapy were deemed RRs and were compared to SRs. Early
virological responses were compared rather than ultimate treatment outcomes to avoid issues
of compliance and treatment tolerance. In addition, we reasoned that the early responses might
be more reflective of altered gene expression by IFN than the ultimate treatment outcome.
There was a good correlation between the early response and late response, with 5 of 6 RRs
achieving an SVR. The other RR was lost to follow-up.
Similarly to the pretreatment samples, the best model for this comparison included gender but
not race. After we controlled for the effect of gender, 2884 genes were differentially regulated
between the RRs and SRs, and 179 genes were detectable in greater than 50% of the samples
and differed by 1.5-fold or more with a P value of less than 0.01. A summary of the differentially
Feld et al. Page 5
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
regulated genes is shown in Table 4, and a full list is available in the supplementary material
(Supplement to Table 4).
No significant differences in the expression of known ISGs were noted between the groups.
Genes involved in IFN-inhibitory pathways were up-regulated in SRs. PIAS1 interacts with
STAT1 and prevents binding to the interferon-sensitive response element (ISRE), resulting in
reduced ISG production.14 The ability of PIAS1 to interact with STAT1 is regulated by the
methylation status of STAT1. PP2A has been reported to interfere with the methylation of
STAT1, thereby increasing PIAS-STAT1 interactions and reducing STAT1-ISRE binding.
PP2A was elevated slowly in comparison with RRs. In addition, ubiquitin-specific peptidase
13 (USP13) was 2.7-fold more highly expressed in SRs. USPs cleave conjugated ubiquitin or
ubiquitin-like molecules from proteins, thus preventing them from being targeted for
degradation in the proteosome. USP18 has specifically been shown to cleave conjugated ISG15
from target proteins. The silencing of USP18 improves IFN responsiveness in vitro, and USP18
was found to be up-regulated in pretreatment liver biopsies from future nonresponders by Chen
et al.6 and in our cohort.15 Whether other USPs have similar IFN inhibitory activity is currently
unknown.
Although the absolute gene expression level is important, the magnitude of gene induction
from the baseline may also be relevant. Because patients could not be biopsied before and
during the treatment, true treatment-related gene induction could not be evaluated. However,
through the use of the pretreatment patients as the baseline for the on-treatment patients, a
surrogate for gene induction was assessed. Thus, RRs in the on-treatment group were compared
to RRs in the pretreatment group, and similarly, SRs were compared in each group. The fold
change was calculated by the division of the mean expression value of the on-treatment group
by the mean expression value of the pretreatment group because even after matching, a direct
comparison would be inappropriate because of interindividual variation in gene expression.
Because the pretreatment group and on-treatment group comprised different patients, this
comparison is not a true measure of gene induction but rather is a surrogate measure of hepatic
gene expressions in response to treatment. Using this comparison, after controlling for gender,
we found that RRs had 873 genes differentially regulated by 1.5-fold or more with a P value
of less than 0.01 between on-treatment and pretreatment groups, whereas SRs had 438 genes
that differed by the treatment group (Table 5 and Supplement to Table 5).
With this surrogate measure, the most striking difference between RRs and SRs was in the
IFN-related genes. Classical ISGs such as Mx1, OAS1-OAS3, ISG15, and viperin had a greater
fold change between on-treatment and pretreatment RRs than SRs, as did proteins involved in
the early IFN cascade such as STAT1. In addition, less well characterized ISGs (ISG20 and
IFN-induced protein 35) and other IFN-related genes (IFN-induced proteins with TTPR 1–5,
IFN transmembrane proteins 1 and 2, guanylate binding protein 1, and 28-kD IFN-responsive
protein) also showed a larger fold change between the on-treatment and pretreatment groups
among RRs. RIG-I and IRF7, which are involved in IFN production through the IRF3/7
pathway, also showed greater fold differences between treatment groups in RRs than SRs.16
This suggests that although the absolute level of expression of ISGs did not differ between the
RRs and SRs, there was a greater fold difference, a surrogate for greater induction of ISGs in
the RRs, and this may potentially account for their improved antiviral response.
In addition to IFN-related genes, several other pathways showed differences in the pretreatment
expression versus the on-treatment expression in a comparison of RRs and SRs. IFN inhibitory
pathways showed a greater fold-change difference in SRs than in RRs. PP2A expression was
6.6-fold higher (P = 0.0001) in on-treatment SRs versus pretreatment SRs but showed no
change among the pretreatment and on-treatment RRs. The small ubiquitin-like modifier
(SUMO) pathway was also affected. Like ubiquitin, SUMO binds to many proteins, targeting
Feld et al. Page 6
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
them for degradation, and it has been shown to bind STAT1, resulting in reduced IFN
responsiveness.17 SUMO-1 was down-regulated in RRs. USP18 showed a greater fold change
in RRs between pretreatment and on-treatment groups, and this likely reflected the fact that
this ISG was significantly up-regulated in the pretreatment SRs.
Some immune-related genes also differed by the fold change between the groups. IP10
[chemokine (C-X-C motif) ligand 10] expression was higher in pretreatment RRs versus on-
treatment RRs, whereas it was lower in on-treatment SRs. In studies of PBMCs, IP10 has been
found to be elevated prior to treatment, with down-regulation during treatment correlating with
viral clearance.18,19 Interleukin-6 was recently reported to be increased in HCV-infected
chimpanzees and was postulated to interfere with IFN signaling possibly through a suppressor
of cytokine signaling 3 (SOCS3)–mediated mechanism.20 Possibly in keeping with this
observation, the expression of the interleukin-6 receptor was markedly lower in on-treatment
RRs versus pretreatment RRs with no change in SRs. Other genes that differed between RRs
and SRs but were of less clear significance include the insulin receptor, the leptin receptor, and
insulin-like growth factor 1.
Ribavirin Versus No Ribavirin
Of the on-treatment patients, 5 received peginterferon alone, whereas 6 were treated with
ribavirin for 72 hours followed by peginterferon, prior to liver biopsy. These 2 groups were
compared to evaluate the independent effects of ribavirin on hepatic gene expression. Neither
race nor gender contributed to the model. A total of 3645 genes were differentially regulated
between the groups, and 563 genes differed by 1.5-fold or greater expression with a P value
of 0.01. A summary of the differentially regulated genes is shown in Table 6, and a full list is
available in the supplementary material (Supplement to Table 6). Gene expression patterns
differed in patients that received ribavirin in 4 main categories with potential relevance to the
treatment response. These included genes with effects on IFN signaling, IFN inhibition, HSC
activation, and apoptosis.
Patients treated with ribavirin and peginterferon showed greater induction of genes involved
in the IFN signaling cascade than those treated with peginterferon alone. The IFN-alpha
receptor was induced 1.4-fold in the ribavirin group, and although this did not meet the
predefined threshold for significance, the P value was highly significant (P = 0.001). This level
of induction early in the cascade may lead to important downstream effects. Similarly, IRF9,
which binds to STAT1 dimers to form IFN-stimulated transcription factor 3, the transcriptional
complex that leads to ISG production by binding to the ISRE,21 was up-regulated in ribavirin-
treated patients 1.3-fold with a P value of 0.0002. IRF7, critical for endogenous IFN production,
was also up-regulated in the ribavirin-treated patients.
In addition, ribavirin had effects on IFN-inhibitory pathways. PP2A was down-regulated in
the ribavirin-treated group; however, although there was a marked reduction in expression by
the fold change (6.8), the P value (0.041) did not reach the 0.01 level of significance. When
evaluated by real-time PCR, PP2A was found to be statistically significantly down-regulated
in the ribavirin-treated patients (Fig. 4D). PP1C was also significantly down-regulated, and
although it has not been shown to directly interfere with STAT1 methylation, given its
homology with PP2A, it is possible that it has similar activity. SOCS1 also interferes with IFN
signaling.22 SOCS1 was down-regulated in the ribavirin-treated patients. Finally, the SUMO
pathway was affected by ribavirin. SUMO1, SUMO3, and the enzyme involved in SUMO
activation, SUMO-activating enzyme 1, were down-regulated by ribavirin; however, SUMO-
specific peptidase 3, which is responsible for SUMO degradation, was also down-regulated in
the ribavirin-treated patients. Overall, ribavirin down-regulated IFN-inhibitory pathways, thus
potentially enhancing the antiviral activity of IFN.
Feld et al. Page 7
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
The HSC is well established as the main cellular orchestrator of hepatic fibrosis.23 In response
to liver injury, the HSC undergoes a phenotypic change from a quiescent cell to a proliferative
myofibroblast-like activated cell that deposits extracellular matrix (ECM) leading to hepatic
fibrosis. Studies have identified numerous triggers for HSC activation. Treatment with ribavirin
led to down-regulation of a number of genes known to promote HSC activation. Transforming
growth factor beta (TGF-β) is one of the most potent stimuli for HSC activation and is also
produced by activated HSC.24 TGF-β1 and TGF-β3, the TGF-β receptor, and TGF-β receptor–
associated protein 1 were all down-regulated in ribavirin-treated patients. Activated HSCs
produce type I collagen.24 Both type I collagen and its receptor CD36 were down-regulated
by ribavirin treatment. Peroxisome proliferator-activated receptor gamma (PPAR-γ) is
expressed in HSC, and upon activation, PPAR-γ expression is reduced.23 In ribavirin-treated
patients, PPAR-γ expression was increased, supporting decreased HSC activation. Matrix
metalloproteinases degrade ECM deposited by HSC but are inhibited by the TIMP family. In
ribavirin-treated patients, matrix metalloproteinase 24 was induced, and TIMP2 was down-
regulated; this favored decreased ECM deposition. Ribavirin treatment resulted in the down-
regulation of genes involved in HSC activation (TGF-β family) and markers of HSC activation
(collagen type 1 and PPAR-γ), thus favoring less HSC activation and potentially less hepatic
fibrosis.
Ribavirin treatment also affected apoptosis pathways. Caspase 8, the main regulator in the
cytoplasmic Fas-associated death domain apoptosis cascade, was induced in ribavirin-treated
patients. Caspase recruitment domain family member 12, an important promoter of apoptosis,
was also up-regulated in the ribavirin group. There was a mix of up-regulation and down-
regulation among other genes affecting apoptosis. On balance, ribavirin treatment appeared to
promote apoptosis; however, given the complexity of the pathways, it is difficult to draw firm
conclusions.
Real-Time PCR Confirmation
In addition to the use of real-time quantitative PCR to confirm the microarray findings in the
original study group, tissue from patients for whom there was inadequate RNA to perform the
microarray was also evaluated. Because the patterns for PCR confirmation were established
only in those for whom the microarray was performed, the additional patients provided a
validation cohort. To increase the numbers, groups were compared by the combination of
patients from both cohorts, and the comparisons were performed within each group
individually as well (Table 1). Not all genes were compared in all patient samples because of
insufficient remaining RNA.
On-treatment patients in both the original and validation cohorts experienced the induction of
STAT1, Mx1, OAS3, ISG15, IRF7, and the IFN-alpha receptor in comparison with untreated
patients. Ribavirin-treated patients showed the induction of ISGs and the IFN-alpha receptor
in comparison with patients that received peginterferon alone. An examination of IFN-
inhibitory pathways showed that PP2A was down-regulated in ribavirin-treated patients.
Markers of HSC activation, including CD36, collagen 1A, and TIMP2, were all down-regulated
in ribavirin-treated patients. Fas was up-regulated in ribavirin-treated patients, and this
suggested increased apoptosis. There was inadequate RNA to evaluate other apoptosis markers.
Representative real-time PCR results are shown in Fig. 4.
Cluster Analysis
With the gene lists generated for each subgroup, cluster analysis was performed and
demonstrated that the groups separated into distinct gene expression groups, as shown in Fig.
5. Separation based on expression profiles was most distinct between pretreatment and on-
treatment patients and between those receiving ribavirin and those treated with peginterferon
Feld et al. Page 8
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
alone. Although patients did separate on the basis of the treatment response in both pretreatment
and on-treatment biopsy samples, the distinctness of the groups was not as great, as evidenced
by closer common branch points.
Discussion
In this study, peginterferon led to the hepatic induction of known ISGs and a large number of
other genes. The list of induced genes was compared to that from published data on IFN-treated
uninfected chimpanzees, primary human hepatocytes, and human PBMCs.7,9 A very similar
pattern of ISG induction was seen. As in PBMCs, transcription factors such as activating
transcription factor 7 and the proapoptotic interleukin-18 pathway were induced in the liver
with peginterferon treatment.9 Genes involved in IFN production, including IRF7 and RIG-I,
were also induced by treatment, and this suggested that therapeutic IFN may also promote
endogenous IFN production. Other pathways induced by treatment included those involved in
the oxidative stress response, apoptosis, and antigen presentation.
Although the lists of gene induced by peginterferon were similar between humans and
chimpanzees, the level of fold induction between treated and untreated patients was lower in
magnitude than the fold induction reported by Lanford et al.7 in chimpanzees. Most classical
ISGs were up-regulated 1.5–3–fold in on-treatment biopsies in comparison with pretreatment
samples, whereas Lanford et al. found induction levels of up to 47-fold. However, differences
in the study design may explain these apparent discrepancies. The chimpanzees were
uninfected at the baseline and served as their own controls. In contrast, in this study, the
pretreatment group comprised HCV-infected individuals. The induction of ISGs from
endogenous IFN is known to occur in chronically infected patients, and the elevation of the
baseline value will greatly reduce the fold induction even if similar absolute levels of gene
expression occur after treatment with IFN.7 With separation by the treatment response, this
difference became more apparent, with greater induction in RRs than SRs, likely because of
lower ISG expression at baseline in the future RRs. The timing of the evaluation may also be
important. Lanford et al. were able to biopsy chimpanzees sequentially and found that after an
initial surge in ISG expression at 4 hours, gene expression quickly dropped off within 24 hours.
Unfortunately, the time course of hepatic gene induction during therapy in naive or infected
humans is unknown, but it may be that there is a similar reduction in gene expression by 24
hours. A comparison of 24-hour gene expression after IFN between humans and chimpanzees
was fairly similar, with the difference in the baseline ISG expression from chronic HCV
infection in humans likely accounting for the differences.
Chen et al.6 previously reported that in pretreatment liver biopsies, nonresponders had higher
hepatic ISG expression than sustained responders or uninfected controls. An examination of
our pretreatment population revealed an identical pattern. All of the ISGs found by Chen et al.
to be differentially regulated were also noted in our analysis. In addition, a number of other
ISGs and IFN regulators, including STAT1 and RIG-I, were found to follow the same pattern
of increased expression in future nonresponders. This result confirms the original findings,
particularly given that not only were different cohorts examined but different microarray
platforms (complementary DNA versus RNA) were also employed. To ensure that our choice
of early virological response was reasonable, we analyzed the data on the basis of both early
and ultimate responses and found similar results (data not shown).
The comparisons of RRs and SRs offer some clues to understanding IFN nonresponse. The
first finding is that absolute ISG expression did not differ significantly between RRs and SRs
on therapy. Coupled with higher pretreatment ISG expression in future SRs, this raises the
question of whether SRs already have maximally induced ISGs and cannot respond further to
therapeutic IFN. To evaluate this issue, we compared ISG expression between SRs before and
Feld et al. Page 9
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
during treatment. For most ISGs, the mean expression level was higher in the on-treatment
SRs; however, this was not true for all ISGs. In addition, the magnitude of the difference was
small, and some individual patients had pretreatment gene expression levels comparable to
those found in on-treatment SRs. In contrast, among RRs, on-treatment ISG expression was
universally higher than pretreatment levels, and the fold change was much greater than that in
SRs. The effect on global gene expression also differed, with almost twice as many genes with
greater than 1.5-fold induction in RRs than SRs. Together, these data suggest that all patients
likely achieve maximal ISG induction on peginterferon, but SRs already have high and possibly
even maximal ISG expression prior to treatment. Although SRs may be able to induce ISGs
further with treatment, they gain little additional benefit from therapy, and this results in a slow
response and ultimately nonresponse.
Although ISG expression was similar between RRs and SRs on therapy, differences were seen
in IFN inhibitory pathways. PP2A levels are higher in patients infected with HCV than in
healthy controls, and both in vitro and in vivo expression of this protein results in
hypomethylation of STAT1, which results in greater interaction with PIAS1, thus reducing
STAT1-ISRE binding and subsequent ISG expression.14 PP2A expression was 8-fold greater
in SRs. Furthermore, in contrast to ISGs, IFN-inhibitory pathways showed a greater change in
the expression level between on-treatment and pretreatment SRs than RRs. The inhibition of
IFN activity may be critical for circumventing the effects of endogenous and therapeutic IFN.
A proposed schematic is shown in Fig. 6.
In contrast to the relative lack of ISG induction in the livers of SRs, ISGs were recently reported
to be strongly induced in PBMC from SRs, albeit somewhat less so than in RRs, after treatment
with IFN.25 The pattern of response to treatment among SRs is very similar to the pattern
recently described in HCV-infected chimpanzees. Huang et al.20 found that like human
nonresponders, chimpanzees had high baseline hepatic but normal PBMC ISG expression
levels. When they were treated with IFN, ISG induction in PBMCs from infected chimpanzees
was found to be at levels only slightly lower than those in naive animals. However, in the liver,
ISG induction was almost completely abrogated in the infected chimpanzees, and little or no
reduction in HCV RNA was seen. As in human SRs, increased expression of IFN-inhibitory
pathways was also seen in the infected animals. The similarities between these patterns of
response suggest that the chimpanzee may serve as a relevant model for understanding IFN
nonresponse.
Despite its clear effectiveness, the mechanisms by which ribavirin improves the response to
IFN are poorly understood. Taylor et al.9 found few differences in gene expression in PBMCs
in patients treated with ribavirin. In contrast, a comparison of hepatic gene expression in
patients receiving ribavirin and peginterferon with those receiving peginterferon alone revealed
differing gene expression patterns, potentially offering some important insights into the
mechanism by which ribavirin affects the treatment response. The most direct effect of ribavirin
was the induction of genes involved in the IFN cascade. In addition to increased expression of
the IFN-alpha receptor, IFN-stimulated transcription factor 3 and IRF7, which promotes
endogenous IFN production, were also induced in ribavirin-treated patients. The effect on IFN
signaling is in keeping with that reported by Zhang et al.,26 who found that ribavirin led to up-
regulation of ISGs and a reporter gene driven by the ISRE promoter in a respiratory syncytial
virus infection. Notably, in this experimental system, ribavirin increased ISRE activity only in
the setting of endogenous IFN production, with no effect seen with ribavirin treatment alone.
This is analogous to the situation seen in the treatment of HCV, in which ribavirin has little
effect as a monotherapy but leads to important synergistic improvements in treatment when
combined with peginterferon. The confirmation of the induction of IFN-alpha receptor and
IRF7 expression by real-time PCR in the cohort not examined by a microarray adds further
weight to the importance of this mechanism. This suggests that ribavirin may contribute to the
Feld et al. Page 10
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
antiviral response by making cells more responsive to IFN through the receptor and furthermore
by increasing the production of endogenous IFN. The down-regulation of IFN-inhibitory
pathways by ribavirin may further enhance the IFN response.
Aside from direct effects on the IFN response, ribavirin treatment altered gene expression in
pathways affecting HSC activation, which may reduce hepatic fibrogenesis. In addition to
known HSC activators, numerous other genes identified by microarray studies of HSC
activation were also down-regulated by ribavirin.23,24 This may account for the finding of
reduced hepatic fibrosis in a subset of patients treated with long-term ribavirin monotherapy.
27 Although the mechanism is unclear, increased hepatic fibrosis is associated with a poorer
treatment response.28,29 Perhaps by reducing fibrogenesis, at least in some patients, ribavirin
further improves IFN efficacy.
Ribavirin also appears to have an effect on apoptotic pathways. Although there were mixed
effects, caspase 8, the main activator of the Fas-mediated apoptosis pathway, was up-regulated
in ribavirin-treated patients. This is in keeping with a previous report from Schlosser et al.,30
who showed that IFN and ribavirin treatment of Hep G2 cells resulted in increased caspase 8
expression and increased Fas-mediated apoptosis. The finding that apoptotic pathways are also
up-regulated in responders to therapy suggests that apoptosis may be a critical component of
the response to HCV treatment.31
This study has some limitations. The sample size in all groups, particularly for ribavirin, is
relatively small; however, it is comparable to the size of many microarray studies. To provide
some validation, the cohort of patients with inadequate RNA for a microarray was evaluated
by real-time PCR with generally confirmatory results. The liver biopsies were performed 24
hours after the first dose of peginterferon. Up-regulation and down-regulation of IFN pathways
occur very rapidly, and even by 24 hours, some important effects may have been missed.7 In
addition, the lack of multiple biopsies also prevents the collection of longitudinal data on gene
expression, which may show important dynamic changes during the course of therapy. Use of
the pre-treatment group as a baseline for the on-treatment group is not ideal, but given the
infeasibility of biopsying patients before and during treatment, it is the best available
alternative. Despite the use of a P value of 0.01 for significance, the possibility of false positive
findings due to multiple comparisons remains. To minimize this issue, a fold-change threshold
was included, and signaling pathways and individual gene expression were also compared.
Finally, important findings were confirmed with real-time PCR.
In summary, we have shown that peginterferon treatment leads to the induction of known ISGs
and many other genes. We have confirmed previous findings showing increased ISG
expression in pretreatment liver biopsies of nonresponders. On treatment, we found that RRs
and SRs have similar ISG expression but SRs have up-regulation of IFN inhibitory pathways.
Evaluating a surrogate for treatment-induced gene induction by controlling for baseline
expression in the pretreatment group, we found that RRs have higher levels of induction of
ISGs, whereas IFN-inhibitory pathways are induced to a greater degree in SRs. Finally,
ribavirin appears to augment the IFN response and down-regulate genes involved in HSC
activation.
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
Supported in part by a grant from Hoffmann-La Roche, by a grant from the General Clinical Research Center of the
University of North Carolina (RR 000046), by a Midcareer Investigator Award in Patient-Oriented Research
Feld et al. Page 11
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
(DK06614; to M.W.F.), Doris Duke Fellowship (LMS) and by the Intramural Research Program of the National
Institute of Diabetes and Digestive and Kidney Diseases (National Institutes of Health).
We thank Karen Dougherty, A.N.P., Roshan Shrestha, M.D., and George Poy for their contributions to this study.
Abbreviations
ECM extracellular matrix
HCV hepatitis C virus
HSC hepatic stellate cell
IFN interferon
IRF interferon regulatory factor
ISG interferon-stimulated gene
ISRE interferon-sensitive response element
Mx myxovirus resistance
OAS oligoadenylate synthetase
PBMC peripheral blood mononuclear cell
PCR polymerase chain reaction
PIAS protein inhibitor of activated signal transducer and activator of transcription
PP2A protein phosphatase 2A
PPAR-γperoxisome proliferator-activated receptor gamma
RIG-I retinoic acid–inducible gene I
RR rapid responder
SOCS suppressor of cytokine signaling
SR slow responder
STAT signal transducer and activator of transcription
SUMO small ubiquitin-like modifier
SVR sustained virological response
TGF-βtransforming growth factor beta
TIMP tissue inhibitor of metallopeptidase
USP ubiquitin-specific peptidase
References
1. Manns MP, McHutchison JG, Gordon SC, Rustgi VK, Shiffman M, Reindollar R, et al. Peginterferon
alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic
hepatitis C: a randomised trial. Lancet 2001;358:958–965. [PubMed: 11583749]
2. Fried MW, Shiffman ML, Reddy KR, Smith C, Marinos G, Goncales FL Jr, et al. Peginterferon alfa-2a
plus ribavirin for chronic hepatitis C virus infection. N Engl J Med 2002;347:975–982. [PubMed:
12324553]
3. Hofmann WP, Zeuzem S, Sarrazin C. Hepatitis C virus-related resistance mechanisms to interferon
alpha-based antiviral therapy. J Clin Virol 2005;32:86–91. [PubMed: 15653410]
Feld et al. Page 12
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
4. Conjeevaram HS, Fried MW, Jeffers LJ, Terrault NA, Wiley-Lucas TE, Afdhal N, et al. Peginterferon
and ribavirin treatment in African American and Caucasian American patients with hepatitis C
genotype 1. Gastroenterology 2006;131:470–477. [PubMed: 16890601]
5. Ferenci P. Predictors of response to therapy for chronic hepatitis C. Semin Liver Dis 2004;24(suppl
2):25–31. [PubMed: 15346243]
6. Chen L, Borozan I, Feld J, Sun J, Tannis LL, Coltescu C, et al. Hepatic gene expression discriminates
responders and nonresponders in treatment of chronic hepatitis C viral infection. Gastroenterology
2005;128:1437–1444. [PubMed: 15887125]
7. Lanford RE, Guerra B, Lee H, Chavez D, Brasky KM, Bigger CB. Genomic response to interferon-
alpha in chimpanzees: implications of rapid downregulation for hepatitis C kinetics. HEPATOLOGY
2006;43:961–972. [PubMed: 16628626]
8. Asahina Y, Izumi N, Uchihara M, Noguchi O, Nishimura Y, Inoue K, et al. Interferon-stimulated gene
expression and hepatitis C viral dynamics during different interferon regimens. J Hepatol
2003;39:421–427. [PubMed: 12927929]
9. Taylor MW, Grosse WM, Schaley JE, Sanda C, Wu X, Chien SC, et al. Global effect of PEG-IFN-
alpha and ribavirin on gene expression in PBMC in vitro. J Interferon Cytokine Res 2004;24:107–118.
[PubMed: 14980075]
10. Lau JY, Tam RC, Liang TJ, Hong Z. Mechanism of action of ribavirin in the combination treatment
of chronic HCV infection. HEPATOLOGY 2002;35:1002–1009. [PubMed: 11981750]
11. Cope L, Hartman SM, Gohlmann HW, Tiesman JP, Irizarry RA. Analysis of Affymetrix GeneChip
data using amplified RNA. Biotechniques 2006;40:165–166. 168, 170. [PubMed: 16526405]
12. Bleicher KB, Pippert TR, Glaab WE, Skopek TR, Sina JF, Umbenhauer DR. Use of real-time gene-
specific polymerase chain reaction to measure RNA expression of three family members of rat
cytochrome P450 4A. J Biochem Mol Toxicol 2001;15:133–142. [PubMed: 11424223]
13. Helbig KJ, Lau DT, Semendric L, Harley HA, Beard MR. Analysis of ISG expression in chronic
hepatitis C identifies viperin as a potential antiviral effector. HEPATOLOGY 2005;42:702–710.
[PubMed: 16108059]
14. Duong FH, Filipowicz M, Tripodi M, La Monica N, Heim MH. Hepatitis C virus inhibits interferon
signaling through up-regulation of protein phosphatase 2A. Gastroenterology 2004;126:263–277.
[PubMed: 14699505]
15. Randall G, Chen L, Panis M, Fischer AK, Lindenbach BD, Sun J, et al. Silencing of USP18 potentiates
the antiviral activity of interferon against hepatitis C virus infection. Gastroenterology
2006;131:1584–1591. [PubMed: 17101330]
16. Foy E, Li K, Wang C, Sumpter R Jr, Ikeda M, Lemon SM, et al. Regulation of interferon regulatory
factor-3 by the hepatitis C virus serine protease. Science 2003;300:1145–1148. [PubMed: 12702807]
17. Ungureanu D, Vanhatupa S, Gronholm J, Palvimo JJ, Silvennoinen O. SUMO-1 conjugation
selectively modulates STAT1-mediated gene responses. Blood 2005;106:224–226. [PubMed:
15761017]
18. Diago M, Castellano G, Garcia-Samaniego J, Perez C, Fernandez I, Romero M, et al. Association of
pretreatment serum interferon gamma inducible protein 10 levels with sustained virological response
to peginterferon plus ribavirin therapy in genotype 1 infected patients with chronic hepatitis C. Gut
2006;55:374–379. [PubMed: 16150856]
19. Lagging M, Romero AI, Westin J, Norkrans G, Dhillon AP, Pawlotsky JM, et al. IP-10 predicts viral
response and therapeutic outcome in difficult-to-treat patients with HCV genotype 1 infection.
HEPATOLOGY 2006;44:1617–1625. [PubMed: 17133471]
20. Huang Y, Feld JJ, Sapp RK, Nanda S, Lin JH, Blatt LM, et al. Defective hepatic response to interferon
and activation of suppressor of cytokine signaling 3 in chronic hepatitis C. Gastroenterology
2007;132:733–744. [PubMed: 17258724]
21. Feld JJ, Hoofnagle JH. Mechanism of action of interferon and ribavirin in treatment of hepatitis C.
Nature 2005;436:967–972. [PubMed: 16107837]
22. Brand S, Zitzmann K, Dambacher J, Beigel F, Olszak T, Vlotides G, et al. SOCS-1 inhibits expression
of the antiviral proteins 23,53-OAS and MxA induced by the novel interferon-lambdas IL-28A and
IL-29. Biochem Biophys Res Commun 2005;331:543–548. [PubMed: 15850793]
Feld et al. Page 13
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
23. Eng FJ, Friedman SL. Transcriptional regulation in hepatic stellate cells. Semin Liver Dis
2001;21:385–395. [PubMed: 11586467]
24. Mann DA, Smart DE. Transcriptional regulation of hepatic stellate cell activation. Gut 2002;50:891–
896. [PubMed: 12010897]
25. Taylor MW, Tsukahara T, Brodsky L, Schaley J, Sanda C, Stephens MJ, et al. Changes in gene
expression during peginterferon and ribavirin therapy of chronic hepatitis C distinguish responders
from non responders to antiviral therapy. J Virol 2007;81:3391–3401. [PubMed: 17267482]
26. Zhang Y, Jamaluddin M, Wang S, Tian B, Garofalo RP, Casola A, et al. Ribavirin treatment up-
regulates antiviral gene expression via the interferon-stimulated response element in respiratory
syncytial virus-infected epithelial cells. J Virol 2003;77:5933–5947. [PubMed: 12719586]
27. Hoofnagle JH, Ghany MG, Kleiner DE, Doo E, Heller T, Promrat K, et al. Maintenance therapy with
ribavirin in patients with chronic hepatitis C who fail to respond to combination therapy with
interferon alfa and ribavirin. HEPATOLOGY 2003;38:66–74. [PubMed: 12829988]
28. Everson GT, Jensen DM, Craig JR, van Leeuwen DJ, Bain VG, Ehrinpreis MN, et al. Efficacy of
interferon treatment for patients with chronic hepatitis C: comparison of response in cirrhotics,
fibrotics, or nonfibrotics. HEPATOLOGY 1999;30:271–276. [PubMed: 10385666]
29. Heathcote EJ, Shiffman ML, Cooksley WG, Dusheiko GM, Lee SS, Balart L, et al. Peginterferon
alfa-2a in patients with chronic hepatitis C and cirrhosis. N Engl J Med 2000;343:1673–1680.
[PubMed: 11106716]
30. Schlosser SF, Schuler M, Berg CP, Lauber K, Schulze-Osthoff K, Schmahl FW, et al. Ribavirin and
alpha interferon enhance death receptor-mediated apoptosis and caspase activation in human
hepatoma cells. Antimicrob Agents Chemother 2003;47:1912–1921. [PubMed: 12760867]
31. Volkmann X, Cornberg M, Wedemeyer H, Lehner F, Manns MP, Schulze-Osthoff K, et al. Caspase
activation is required for antiviral treatment response in chronic hepatitis C virus infection.
HEPATOLOGY 2006;43:1311–1316. [PubMed: 16729308]
Feld et al. Page 14
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
Study design. The patients in the on-treatment group were given either 180 μg of peginterferon-
alpha 2a subcutaneously 24 hours prior to liver biopsy or 1000/1200 mg of ribavirin daily for
72 hours plus 180 μg of peginterferon-alpha 2a subcutaneously 24 hours prior to liver biopsy.
After the biopsy, the patients were continued on the therapy for 48 weeks.
Feld et al. Page 15
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Heat map showing distinct gene expression patterns in pretreatment and on-treatment groups.
The map is based on the 364 genes with a greater than 1.5-fold difference in the expression
with P <0.01. Red indicates increased gene expression, and blue indicates decreased gene
expression. The white line across and down the heat map provides a statistically significant
separation of the 2 groups and up-regulated or down-regulated genes. Within the 2 groups, the
treatment response is indicated as follows: N, naive treatment; RR, rapid responder; and SR,
slow responder.
Feld et al. Page 16
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 3.
Gene pathways with significant differential gene expression between pretreatment and on-
treatment patients. For each known pathway, the number of genes with differing expression
between the groups was calculated, and a P value was determined on the basis of the likelihood
of finding the given number of genes by chance alone. The inverse logarithm of the P value is
shown on the x axis. Pathways with a cutoff of 2 (equivalent to a P value of 0.01) are shown.
In addition to interferon signaling, pathways involved in immune responses and apoptosis were
most affected by the interferon treatment.
Feld et al. Page 17
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 4.
Analyses of the gene expression by a quantitative polymerase chain reaction. Selected genes
from the microarray analysis were quantified by a TaqMan real-time polymerase chain
reaction. (A) On-treatment versus pretreatment. (B) Future RRs versus SRs (pretreatment
group). (C) RRs versus SRs (on-treatment group by the fold change). Gene expression in the
on-treatment group was normalized for the baseline expression with the mean expression level
in the pretreatment group. The fold change was calculated as on-treatment RRs/pretreatment
RRs or on-treatment SRs/pretreatment SRs. (D) Peginterferon and ribavirin versus
peginterferon. RR indicates rapid responder; and SR, slow responder.
Feld et al. Page 18
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 5.
Supervised hierarchical clustering by a gene list. The patients are categorized on the basis of
gene expression profiles. The number of branch points between patients reflects the degree of
similarity in the expression pattern. (A) On-treatment group versus the pre-treatment group.
(B) Future slow responders versus rapid responders in the pretreatment group. (C) Rapid
responders versus slow responders in the on-treatment group divided by the pretreatment
group. (D) Peginterferon plus ribavirin versus peginterferon alone.
Feld et al. Page 19
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 6.
Postulated scheme of the importance of gene expression before and during peginterferon and
ribavirin treatment and its effect on treatment outcome. The strength of the adaptive immune
response and early endogenous IFN production likely determine whether the initial HCV
infection is cleared or becomes chronic. High ISG expression, presumably from increased
endogenous IFN production, is accompanied by up-regulation of IFN-inhibitory pathways.
This leads to reduced ISG induction with IFN treatment. Ribavirin enhances the efficacy of
IFN directly through up-regulation of the IFN receptor and indirectly through effects on
apoptosis and possibly HSCs. Ultimately, the degree of ISG induction with therapy may
determine treatment outcome. HCV indicates hepatitis C virus; HSC, hepatic stellate cell; IFN,
interferon; ISG, interferon-stimulated gene; RR, rapid responder; and SR, slow responder.
Feld et al. Page 20
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feld et al. Page 21
Table 1
Baseline Characteristics of the On-Treatment and Pretreatment Groups Used for Microarrays and Quantitative
Real-Time PCR
Microarray Only Microarray and Real-Time PCR
On-Treatment
Group (n = 11) Pretreatment
Group (n = 19) On-Treatment
Group (n = 21) Pretreatment
Group (n = 31)
Gender (male/female) 5/6 10/9 13/8 19/12
Age 45.7 ± 6.9 51.7 ± 4.5 44.9 ± 5.3 51.6 ± 6.0
Ethnicity
African American 6 8 9 14
Caucasian 5 11 12 17
Treatment response
RR 6 5 10 10
SR 5 10 11 14
Naive NA 4 NA 7
Baseline HCV RNA
>6 log copies/mL 6 6 13 16
<6 log copies/mL 5 13 8 15
Biopsy stage*
03 3 7 4
15 10 10 18
23 1 3 4
30 5 1 5
40 0 0 0
Biopsy grade*
00 0 0 0
13 5 9 10
28 9 12 15
30 5 0 6
40 0 0 0
*The biopsy stages and grades follow the Batts and Ludwig scoring system. For RRs, there was a 2 log copies/mL decline in HCV RNA by 4 weeks
of treatment. For SRs, there was a <2 log copies/mL decline in HCV RNA by 4 weeks of treatment. Abbreviations: HCV, hepatitis C virus; NA, not
applicable; PCR, polymerase chain reaction; RR, rapid responder; SR, slow responder.
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feld et al. Page 22
Table 2
Selected Genes Induced by Peginterferon Treatment with a Fold Change Greater Than or Equal to 1.5 (P 0.01)
Gene Category Gene Name Gene Symbol Fold Change (On-Treatment/Pretreatment) P
ISG 2,5-Oligoadenylate synthetase 3 OAS3 2.2 0.0001
Adenosine deaminase, RNA-specific ADAR 1.5 2.6 × 107
Guanylate binding protein 1 GBP1 2.3 9.2 × 106
IFN transmembrane protein 1 (9–27) IFITM1 1.7 0.0009
IFN-alpha–inducible (ISG 15) ISG15 1.8 0.001
IFN-induced protein 35 IFI35 1.7 0.0001
IFN-induced protein 44 IFI44 2.4 3.8 × 105
IFN-induced protein with TTPR 3 IFIT3 1.7 0.004
IFN-induced protein with TTPR 5 IFIT5 1.5 3.1 × 105
Myxovirus resistance 1 Mx1 2.3 9.2 × 106
Myxovirus resistance 2 Mx2 2.5 5.9 × 107
Phospholipid scramblase 1 PLSCR1 3.0 4.6 × 108
Ubiquitin specific protease 18 USP18 1.8 0.0009
Viperin RSAD2 2.3 5.3 × 105
Apolipoprotein B messenger RNA
editing enzyme catalytic polypeptide-3A APOBEC3A 3.8 6.9 × 108
IFN-related IFN-alpha/beta receptor 2 IFNAR2 1.7 0.0001
IFN regulatory factor 7 IRF7 2.5 6.0 × 105
RIG-I DEAD box polypeptide 58 DDX58 1.6 0.0018
Signal transducer/activator transcription STAT1 2.3 1.2 × 106
Immune Beta-2-microglobulin B2M 2.3 3.2 × 105
Chemokine ligand 8 CCL8 4.6 1.16 × 108
Chemokine ligand 19 CCL19 2.6 0.005
C-reactive protein CRP 4.0 0.0032
Interleukin 18 binding protein IL18BP 1.8 2.6 × 105
Interleukin 6 signal transducer IL6ST 2.3 7.5 × 106
Major histocompatibility complex F HLA-F 1.5 0.0002
N-myc and STAT interactor NMI 1.7 2.8 × 106
Nuclear factor of activated T cells NFAT5 1.6 0.0002
Serum amyloid A1 SAA1 10.5 7.0 × 105
Abbreviations: IFN, interferon; ISG, interferon-stimulated gene.
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feld et al. Page 23
Table 3
Selected Genes Differing Between Future RRs and SRs with a Fold Change Greater Than or Equal to 1.5 (P
0.01) in the Pretreatment Group
Gene Category Gene Name Gene Symbol Fold Change (RR/SR) P
ISG 2,5-Oligoadenylate synthetase 2*† OAS2 2.4 0.02
2,5-Oligoadenylate synthetase 3*† OAS3 2.4 0.01
IFN-induced transmembrane protein 1 IFITM1 2.3 0.001
IFN-induced transmembrane protein 3 IFITM3 1.5 0.004
IFN-alpha–inducible protein (ISG-15)*† G1P2/ISG15 3.2 0.028
IFN-alpha–inducible protein (IFI-6–16)G1P3/IFI6 3.0 0.0016
IFN-alpha–inducible protein 27 IFI27 3.7 0.004
IFN-induced protein 35 IFI35 2.3 0.0048
IFN-induced protein 44 IFI44 2.7 0.001
IFN-induced protein with TTPR 1IFIT1 2.8 0.01
IFN-induced protein with TTPR 3 IFIT3 2.6 0.0075
Myxovirus resistance 1MX1 3.9 0.0004
Phospholipid scramblase 1 PLSCR1 2.0 0.003
Ubiquitin-specific peptidase 18*† USP18 1.7 0.042
ViperinRSAD2 3.3 0.0008
IFN-Related RIG-I DEAD box polypeptide 58 DDX58 2.0 0.01
Signal transducer activator of
transcription 1 STAT1 2.0 0.0048
Other Activating transcription factor 7
interacting protein 2ATF7IP2 1.7 0.0083
Chemokine ligand 9 (MIG) CXCL9 2.9 0.0062
Leucine aminopeptidase 3*† LAP3 1.5 0.030
Ribosomal protein S28RPS28 1.98 0.0077
Syntaxin binding protein 5-likeSTXBP5L 6.4 0.0003
*P between 0.05 and 0.01.
Gene identified by Chen et al.6 as differing between future sustained responders and future nonresponders. Abbreviations: IFN, interferon; ISG,
interferon-stimulated gene.
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feld et al. Page 24
Table 4
Direct Comparison of Selected Genes Differing Between RRs and SRs with a Fold Change Greater Than or Equal
to 1.5 (P 0.01) in the On-Treatment Group
Gene Category Gene Name Gene Symbol Fold Change (RR/SR) P
IFN-inhibitory Protein phosphatase regulatory
subunit (formerly 2A) PPP2R3A 8.4 0.0004
Protein phosphatase 3 catalytic
subunit (formerly 2B) PPP3CB 2.4 0.0005
Ubiquitin-specific peptidase 13 USP13 2.7 3.0 × 105
Other Insulin-like growth factor 1 IGF1 2.7 0.0025
Abbreviations: IFN, interferon; RR, rapid responder; SR, slow responder.
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feld et al. Page 25
Table 5
Comparison of a Surrogate for the Fold Induction of Selected Genes Differing Between RRs and SRs with a Fold Change Greater Than or Equal to 1.5 (P
0.01) in the On-Treatment Group Versus the Pretreatment Group (On-Treatment RRs/Pretreatment RRs versus On-Treatment SRs/Pretreatment SRs)
RRs (On-Treatment/Pretreatment) SRs (On-Treatment/Pretreatment)
Gene Category Gene Name Gene Symbol Fold Change PFold Change P
ISG 2,5-Oligoadenylate synthetase 1 OAS1 6.7 0.0004 1.0 NS
2,5-Oligoadenylate synthetase 2 OAS2 16.5 2.9 × 1051.2 NS
2,5-Oligoadenylate synthetase 3 OAS3 8.8 0.0005 1.2 NS
28-kD IFN-responsive protein IFRG28 3.5 0.0003 NC NS
Adenosine deaminase, RNA-specific ADAR 1.8 1.5 × 106NC NS
Guanylate binding protein 1 GBP1 2.5 0.0003 NC NS
IFN-induced transmembrane protein 1 (9–27) IFITM1 5.4 0.0063 1.1 NS
IFN-induced transmembrane protein 2 IFITM2 1.7 0.0004 NC NS
IFN-induced with helicase C domain IFIH1 3.4 1.3 × 105NC NS
IFN-stimulated exonuclease gene 20Kda ISG20 4.6 0.0014 NC NS
IFN-induced protein 35 IFI35 4.6 0.0002 1.1 NS
IFN-induced 44-like IFI44L 57.9 0.0066 1.3 NS
IFN-induced protein with TTPR 1 IFIT1 6.0 0.0003 NC NS
IFN-induced protein with TTPR 2 IFIT2 5.9 0.0022 NC NS
IFN-induced protein with TTPR 3 IFIT3 5.8 0.0005 1.1 NS
IFN-induced protein with TTPR 5 IFIT5 3.0 0.0001 1.1 NS
IFN-stimulated gene 15 ISG15 13.6 0.005 1.6 0.014*
Myxovirus resistance 1 Mx1 17.7 0.006 1.1 NS
Myxovirus resistance 2 Mx2 5.7 2.7 × 1051.6 NS
Viperin RSAD2 12.1 0.0007 1.9 1.1 × 105
IFN-related IFN-alpha 4 IFNA4 3.4 0.0070 NC NS
IFN receptor IFNAR2 NC NS 2.4 0.0011
IFN regulatory factor 7 IRF7 3.3 0.0040 1.2 NS
IFN-stimulated transcription factor ISGF3G 2.0 1.5 × 106NC NS
RIG-I DEAD box polypeptide 58 DDX58 2.7 5.8 × 107NC NS
Signal transducer activator of transcription 1 STAT1 5.2 0.0001 1.8 0.003
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feld et al. Page 26
RRs (On-Treatment/Pretreatment) SRs (On-Treatment/Pretreatment)
Gene Category Gene Name Gene Symbol Fold Change PFold Change P
IFN-inhibitory Protein inhibitor of activated STAT 2 PIAS2 NC NS 1.1*0.00001
Protein inhibitor of activated STAT 4 PIAS4 NC NS 1.2*5.4 × 105
Protein phosphatase 2 regulatory subunit B PPP2R2C NC NS 3.5 0.0002
gamma (formerly 2A)
Protein phosphatase 2 regulatory subunit B PPP2R3A NC NS 6.6 0.0001
alpha (formerly 2A)
Small ubiquitin-like molecule 1 SUMO1 1.6 0.006 NC NS
Ubiquitin-specific peptidase 13 USP13 NC NS 2.3 0.0002
Ubiquitin-specific peptidase 18 USP18 5.33 2.5 × 106NC NS
Immune Chemokine (C-X-C motif) receptor 1 CCR1 2.0 0.0023 NC NS
Chemokine (C-C motif) ligand 14 CCL14 1.6 0.0002 2.8 0.0001
Chemokine (C-X-C motif) ligand 11 CXCL11 5.4 0.0008 NC NS
Complement component 1 subunit s1 C1S 7.1 3.9 × 106NC NS
Complement component 2 C2 2.0 0.0041 NC NS
Complement component 9 C9 1.5 0.0013 NC NS
Interleukin 6 receptor IL6R 6.4 9.0 × 107NC NS
Interleukin 13 receptor alpha 1 IL13RA1 2.0 3.9 × 1051.1 NS
Interleukin 17 receptor B IL17RB 2.1 0.0007 2.1 2.7 × 106
Interleukin 18 binding protein IL18BP 2.7 2.8 × 105NC NS
Other Insulin receptor INSR 2.0 2.4 × 105NC NS
Insulin-like growth factor 2 IGF2 1.7 1.3 × 105NC NS
Integrin beta 1 ITGB1 2.3 2.7 × 107NC NS
Leptin receptor LEPR 3.8 4.1 × 1061.3 NS
*P between 0.05 and 0.01 or a fold change between 1.0 and 1.5. Abbreviations: IFN, interferon; ISG, interferon-stimulated gene; NC, no change in the gene expression detected between groups; NS, not
statistically significant; RR, rapid responder; SR, slow responder.
Hepatology. Author manuscript; available in PMC 2010 January 19.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Feld et al. Page 27
Table 6
Selected Genes Induced by Ribavirin with a Fold Change Greater Than or Equal to 1.5 (P 0.01) in the On-
Treatment Group
Gene Category Gene Name Gene Symbol
Fold Change
(Ribavirin/
Peginterferon
Alone) P
IFN-related IFN-alpha/beta receptor 2*IFNAR2 1.4 0.0012
IFN regulatory factor 7*IRF-7 1.7 0.035
IFN-stimulated transcription factor 3*ISGF3 1.3 0.019
Janus kinase 1 JAK1 2.8 0.0009
IFN-inhibitory Protein phosphatase 2A*PP2CA 6.8 0.042
Protein phosphatase 3 catalytic subunit
(formerly 2B) PPP3CA 1.8 0.003
Small ubiquitin-like molecule 1*SUMO1 1.7 0.031
SUMO-1 activating enzyme*SAE1 1.7 0.048
SUMO-1 peptidase 3 SENP3 2.8 0.0065
Suppressor of cytokine signaling 1*SOCS1 1.7 0.035
HSC CD 36 (collagen I receptor)*CD36 2.7 0.047
Collagen type I alpha 2*COL1A2 1.5 0.021
CREBBP/EP 300 inhibitor 1*CRI1 2.0 0.027
Latent TGF-β binding protein 2*LTBP2 1.4 0.0066
Kruppel-like factor 9 KLF9 1.9 0.0033
Matrix metallopeptidase 24 MMP24 4.2 0.0026
Peroxisome proliferator-activated
receptor gamma PPARGC1B 1.6 0.01
SMAD mothers against DPP homolog 4 SMAD4 1.7 0.01
Tissue inhibitor of metallopeptidase 2*TIMP2 1.7 0.031
Transforming growth factor beta 3 TGFB3 3.2 0.0050
TGF-β receptor–associated protein TGFBRAP1 3.9 0.0027
Apoptosis Apoptosis caspase activation inhibitor*AVEN 5.7 0.013
Apoptosis inhibitor 15*AP15 1.7 0.032
Caspase 8*CASP8 1.8 0.047
BCL-2 associate athanogene BAG2 2.8 0.0047
Caspase recruitment domain family 12 CARD12 4.1 0.010
P53-regulated apoptosis-inducing protein P53AIP1 4.9 0.01
Programmed cell death 5*PDCD5 1.3 0.005
Serine/threonine kinase 17b STK17B 3.9 0.01
TRAF2 and NCK interacting kinase TNIK 6.5 0.0078
*P between 0.05 and 0.01 or a fold change between 1.0 and 1.5. Abbreviations: HSC indicates hepatic stellate cell; and IFN, interferon.
Hepatology. Author manuscript; available in PMC 2010 January 19.
... In our study, as expected the use of injecting drugs was frequently associated with HCV infection among HIV positive patients and HCV transmission was attributable to the use of intravenous drug in almost two third of the cases. This is in line with other studies from Vietnam, Asia and South East Asia that reported a high prevalence of HCV infection, up to 96% in IDUs [9,10,29]. In Hanoi, among HIV-infected individuals, 35.4% were tested positive for HCV and HIV-HCV co-infection was mainly observed in young males and use of injecting drug was reported as a transmission factor in 44% of HCV positive cases [21]. ...
... 19 Circulating IP-10 concentration is well correlated with intrahepatic IP-10 messenger RNA expression during chronic HCV infection. 29 The high baseline hepatic IFN-stimulated genes expression is associated with a lower chance to be SVR to IFN-based therapy when HIV-uninfected. [30][31][32][33] Interestingly, the pre-treatment MCP-1 concentration was also found at a higher concentration in the NR group. ...
Thesis
Les hépatites virales constituent un enjeu majeur de santé public dans le domaine des maladies infectieuses. L’enjeu pour les prochaines années sera de mettre en œuvre des stratégies de dépistage et de de prise en charge des hépatites virales et du VIH, en particulier chez les usagers de drogue par voie intraveineuse et dans les pays à ressources limitées. Dans ce travail de thèse nous avons évalué des outils et des stratégies nouvelles pour améliorer le diagnostic et la prise en charge des infections par le VIH et le VHC dans les pays du Sud. Notre première étude porte sur l’évaluation de la performance diagnostique de la primo-infection du VIH sur Dried serum spot (DSS) par rapport aux tests rapides. Cette étude a été rétrospectivement réalisée chez 39 cas diagnostiqué Primo-infection du VIH entre 2000 et 2012 au CHU de Montpellier. En comparant avec le résultat sur le DSS, nous avons réussi à avoir une sensibilité de 87.2% (34/39) avec PCR Ag/Ac combiné 4eGeneration (ElecsysCombi PT test reactive et Liaison XL test reactive) témoignant de la bonne performance de ce test. Nous avons réalisé par la suite une étude clinico-virologique chez 104 patients co-infectés par le VIH et le VHC à Haiphong, Vietnam (ANRS 12262). C’est pour la première fois qu’une telle étude descriptive (évaluation clinico-virologique, évaluation de la fibrose hépatique avec les marqueurs biologiques recommandés par l’OMS et également par le Fibroscan®) est réalisée dans cette population au Vietnam. L’évaluation clinique, l’analyse des tests biochimiques et virologique et celle de la fibrose par l’élastographie hépatique (Fibroscan®, Echosens) ont été réalisées Parmi 104 patients, 93 (89,4%) avaient des ARN du VHC détectable (médiane 6,19 (4,95 à 6,83 log10 UI / mL). La plupart des patients ont été infectés par les génotypes 1a/1b (69%) et les génotypes 6a/6e (26%). 43 patients (41,3%) présentaient une fibrose significative (≥ F2), dont 24 patients (23,1%) avec une fibrose extensive (F3) et/ou une cirrhose hépatique (F4). L’évaluation de la fibrose hépatique par l’élastométrie (Fibroscan®) est utilisée comme l’examen de référence à comparer avec le test APRI (AST-to-platelet ratio index) recommandé par l’OMS dans la prise en charge des patients infectés par le VHC dans les pays à ressource limitée, en prenant le cut-off élevé supérieur ou égale à 2, nous avons réussi à classifier les fibroses sévères/cirrhose hépatique avec une sensibilité à 90% et une spécificité à 84% (Se: 90%, Sp: 84%, AUROC = 0,93, IC 95%: 0,86 à 0,99).Notre dernière étude a évalué la réponse des marqueurs immunologiques à l’association de Peginterféronα (PegIFNα) et ribavirine. Nous avons étudié la concentration des 25 cytokines chez 30 patients co-infectés par le VIH et le VHC. Les résultats ont été comparés entre les patients classés « RVS » (réponse virologique soutenue) (RVS, n = 19) et les non-répondeurs (NR, n = 11). Le changement en concentration des cytokines lié à la thérapie PegIFNα-ribavirine a été observé chez les patients RVS et NR. Le changement en concentration sérique de l'IL-8, MIP-1β; et MCP-1 pourrait être associé à l'efficacité de la bithérapie PegIFNα-ribavirine chez les patients co-infectés par le VIH et le VHC.L’amélioration du dépistage et du diagnostic est un élément primordial faisant partie du contrôle de ces infections virales. Notre étude a renforcé l’hypothèse de l’utilité de DBS. Le Fibroscan® et les marqueurs biologiques (APRI Test) sont des outils intéressants pour l’évaluation de la fibrose hépatique dans les pays à ressources limitées. Mot clés : Co-infection HIV-VHC, usagers de drogues par voie intraveineuse, dried blood Ssot, fibrose hépatique, fibroscan, APRI, interféron pégylé, Vietnam.
... However, an in vitro experimental study has demonstrated that the combination of ribavirin and INFα1b promoted higher cell survival rates compared to ribavirin alone. The inhibitory effect of the combination was achieved at lower concentrations than the individual drugs alone at higher concentrations, suggesting synergistic effects (32). Additionally, the combination of ribavirin and INFα1b resulted in the upregulation of signal transducer and activator of transcription (STAT) 1 and STAT2, IFN regulatory factors (IRF) 9, and ISG 15 expression, as well as increased phosphorylation levels of STAT1 and STAT2, indicating the potential involvement of ribavirin in the janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway (33). ...
Article
Full-text available
Background: Human adenovirus (HAdV) infections in children can lead to profound pulmonary injury and are frequently associated with severe complications, particularly in cases concomitant with plastic bronchitis. Managing this condition presents significant challenges and carries an exceptionally high fatality rate. Regrettably, there are currently no specific antiviral agents that have demonstrated efficacy in treating severe adenovirus pneumonia in children. Case presentation: We report a 10-month-old infant suffering from severe adenovirus pneumonia combined with plastic bronchitis (PB). He received intravenous ribavirin combined with recombinant human interferon α1b (INFα1b) aerosol inhalation and his condition eventually improved. No side effects occurred during the treatment, and the long-term prognosis was favorable. Conclusion: In this case, the combination therapy of intravenous ribavirin and INFα1b seems to have contributed to the resolution of illness and may be considered for similar cases until stronger evidence is generated.
... However, an in vitro experimental study has demonstrated that the combination of ribavirin and INFα1b promoted higher cell survival rates compared to ribavirin alone. The inhibitory effect of the combination was achieved at lower concentrations than the individual drugs alone at higher concentrations, suggesting synergistic effects (32). Additionally, the combination of ribavirin and INFα1b resulted in the upregulation of signal transducer and activator of transcription (STAT) 1 and STAT2, IFN regulatory factors (IRF) 9, and ISG 15 expression, as well as increased phosphorylation levels of STAT1 and STAT2, indicating the potential involvement of ribavirin in the janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway (33). ...
Article
Full-text available
Background Human adenovirus (HAdV) infections in children can lead to profound pulmonary injury and are frequently associated with severe complications, particularly in cases concomitant with plastic bronchitis. Managing this condition presents significant challenges and carries an exceptionally high fatality rate. Regrettably, there are currently no specific antiviral agents that have demonstrated efficacy in treating severe adenovirus pneumonia in children. Case presentation We report a 10-month-old infant suffering from severe adenovirus pneumonia combined with plastic bronchitis (PB). He received intravenous ribavirin combined with recombinant human interferon α1b (INFα1b) aerosol inhalation and his condition eventually improved. No side effects occurred during the treatment, and the long-term prognosis was favorable. Conclusion In this case, the combination therapy of intravenous ribavirin and INFα1b seems to have contributed to the resolution of illness and may be considered for similar cases until stronger evidence is generated.
... Serum CXCL-10 is highly correlated with intrahepatic CXCL-10 mRNA expression [19] and is often used as a surrogate marker of hepatic ISG expression [9] . Thus, increased serum CXCL-10 and elevated pretreatment ISG expression are both predictors of nonresponse to pegylated IFN-based treatment [22,23] . ...
... Monitoring the ectopic expression of ISGs is another instrumental way to identify ISGs that are individually sufficient for viral suppression [26], for example, interferon-stimulated exonuclease gene 20 (ISG20) and ISG15 ubiquitin-like modifier (ISG15). Using RNA sequencing [27] and fold change-based criteria to measure whether a target human gene is induced by IFN signalling is routinely used [24,28,29]. In most cases, a gene is defined as IFN stimulated (upregulated) when its expression value is increased in the presence of IFNs (fold change >2) [3,24,30]. ...
Article
Full-text available
Background A virus-infected cell triggers a signalling cascade, resulting in the secretion of interferons (IFNs), which in turn induces the upregulation of the IFN-stimulated genes (ISGs) that play a role in antipathogen host defence. Here, we conducted analyses on large-scale data relating to evolutionary gene expression, sequence composition, and network properties to elucidate factors associated with the stimulation of human genes in response to IFN-α. Results We find that ISGs are less evolutionary conserved than genes that are not significantly stimulated in IFN experiments (non-ISGs). ISGs show obvious depletion of GC content in the coding region. This influences the representation of some compositions following the translation process. IFN-repressed human genes (IRGs), downregulated genes in IFN experiments, can have similar properties to the ISGs. Additionally, we design a machine learning framework integrating the support vector machine and novel feature selection algorithm that achieves an area under the receiver operating characteristic curve (AUC) of 0.7455 for ISG prediction. Its application in other IFN systems suggests the similarity between the ISGs triggered by type I and III IFNs. Conclusions ISGs have some unique properties that make them different from the non-ISGs. The representation of some properties has a strong correlation with gene expression following IFN-α stimulation, which can be used as a predictive feature in machine learning. Our model predicts several genes as putative ISGs that so far have shown no significant differential expression when stimulated with IFN-α in the cell/tissue types in the available databases. A web server implementing our method is accessible at http://isgpre.cvr.gla.ac.uk/. The docker image at https://hub.docker.com/r/hchai01/isgpre can be downloaded to reproduce the prediction.
... When RBA is used in conjunction with pegylated interferon-α (peg-IFN) in the treatment of HCV, efficacy of the latter has been shown to be greatly improved, although the mechanisms by which this occurs have not been well defined (356) . However, there have been a number of proposed mechanisms of action for RBA including the upregulation of interferon stimulated gene (ISG) expression, inhibition of host inosine monophosphate dehydrogenase (IMPDH), lethal mutagenesis when ribavirin triphosphate (RTP) is misincorporated into nascent viral genomes, and chain termination during viral RNA synthesis (357)(358)(359) . ...
Thesis
Full-text available
The life cycle of HCV involves a dysregulation in host lipid metabolism that may precede metabolic complications such as insulin resistance (IR), type 2 diabetes mellitus (T2DM), and hepatic steatosis which are often associated with chronic infection. Thioredoxin-Interacting Protein (TXNIP), a host factor that is induced by oxidative stress, and associated with disturbances in host lipid and glucose metabolism, has been reported to be overexpressed during HCV infection, whereby its siRNA-mediated knockdown resulted in a significant impairment of virus replication. This study investigated the mechanisms by which HCV may bring about the overexpression of TXNIP by examining the unfolded protein response (UPR) and mitochondrial dysfunction in HCV gt2a infection models. These pathways were tested in HCV SGR- and JFH-1 infected Huh7 cells by targeting inositol-requiring enzyme 1α (IRE-1α) activity and mitochondrial voltage-dependent anion channel (VDAC) oscillation, and the effects on TXNIP expression, lipid metabolism, and virus replication were measured. The data from this work indicate that the UPR is more relevant to HCV SGR replication, whilst ameliorating mitochondrial dysfunction appears to restore glucose metabolism, downregulate lipogenic gene transcription, inhibit lipid droplet (LD) biogenesis, and reduce JFH-1 RNA replication.
... The drug is thought to function via direct (interference with RNA capping, polymerase inhibition, lethal mutagenesis) and indirect (immunomodulatory effects, inhibition of inosine monophosphate dehydrogenase) mechanisms [12]. It has also been suggested that the drug could increase the activity of IFN-sensitive target genes [14]. Like interferon, ribavirin had been available for medical use prior to the discovery of HCV [15]. ...
Article
Full-text available
Remarkable scientific breakthroughs have been made in the stride towards the development of potent and tolerable hepatitis C regimens within the last three decades. Earlier approaches involved the use of pegylated interferon alfa and ribavirin as standard-of-care treatment. Treating genotype 1a infection with this regimen which was at that time considered the gold standard for hepatitis C virus therapy was rife with challenges; safety and toxicity issues necessitated a rigorous quest for alternative regimens. Deeper understanding of the pathogenesis of hepatitis C virus ushered in the era of direct acting antiviral agents. These agents have been the subject of intensive research in the last two decades, leading to the development of drug classes such as protease inhibitors (e.g., grazoprevir), NS5A inhibitors (e.g., daclatasvir) and NS5B inhibitors (e.g., sofosbuvir). While many are still under development, several have been approved for hepatitis C therapy. A number of studies investigating the combination of direct acting antiviral agents with or without pegylated interferon and/or ribavirin for the treatment of chronic hepatitis have demonstrated sustained virologic response of > 90%. Given the array of direct acting antiviral agents currently available, the present landscape of hepatitis C therapy is now characterized by a gradual transition to all-oral interferon-free regimens. Despite these milestones, the WHO global target of eliminating hepatitis C as a public health problem by 2030 seems uncertain. In this review, we provide a concise account of the evolution and advancements in the development of anti-HCV regimens.
... Although Interferon (IFN) is the one of the proven efficacy in the treatment of hepatitis C, though its mechanisms of treatment failure are inadequately understood, earlier reports have proposed IFN-stimulated genes and the inability to develop effective anti-HCV immunity as possible explanations [11]. In liver diseases, especially in non-alcoholic steatohepatitis, the effect of impaired peroxisomal polyunsaturated fatty acid (PUFA) metabolism and non-enzymatic oxidation on FA constitution is associated with disease progression [12]. ...
Article
Full-text available
Background: Ribavirin is a broad-spectrum nucleoside antiviral drug with multimodal mechanisms of action, which supports its longevity and quality as a clinical resource. It has been widely administered for measles and coronavirus infections. Despite the large amount of data concerning the use of ribavirin alone or in combination for measles, severe acute respiratory syndrome, Middle East respiratory syndrome, and coronavirus disease 2019 (COVID-19) outbreaks, the conclusions of these studies have been contradictory. Underlying reasons for these discrepancies include possible study design inaccuracies and failures and misinterpretations of data, and these potential confounds should be addressed. Aim: To determine the confounding factors of ribavirin treatment studies and propose a therapeutic scheme for COVID-19. Methods: PubMed database was searched over a period of five decades utilizing the terms "ribavirin" alone or combined with other compounds in measles, severe acute respiratory syndrome, Middle East respiratory syndrome, and COVID-19 infections. The literature search was performed and described according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Articles were considered eligible when they reported on ribavirin dose regimens and/or specified outcomes concerning its efficacy and/or possible adverse-effects. In vitro and animal studies were also retrieved. A chapter on ribavirin's pharmacology was included as well. Results: In addition to the difficulties and pressures of an emerging pandemic, there is the burden of designing and conducting well-organized, double-blind, randomized controlled trials. Many studies have succumbed to specific pitfalls, one of which was identified in naturally ribavirin-resistant Vero cell lines in in vitro studies. Other pitfalls include study design inconsistent with the well-established clinical course of disease; inappropriate pharmacology of applied treatments; and the misinterpretation of study results with misconceived generalizations. A comprehensive treatment for COVID-19 is proposed, documented by thorough, long-term investigation of ribavirin regimens in coronavirus infections. Conclusion: A comprehensive treatment strictly tailored to distinct disease stages was proposed based upon studies on ribavirin and coronavirus infections.
Article
Full-text available
Background and Objective With the initial case of corona reported from Wuhan, China on 31st December 2020, there has been an unprecedented rise in the coronavirus disease (COVID-19), with over 200 countries all across the world in less than 3 months. By the October 2020, about 40 million population of the world got infected and over one million deaths occurred. Since no WHO and FDA approved medications or vaccines for COVID-19 were available, there was an impatient bustling need to develop a drug for the treatment. Drug repurposing emerged as the easiest and fast emerging strategy to get medicine for COVID-19 with rapid approvals for the clinical trials. The purpose of this study was to evaluate the status of drug repurposing under the clinical and its impact for the development of medicine for COVID-19. Methodology The study was undertaken to review various clinical trials from www.clinicaltrials.gov website. We evaluated 220 ongoing clinical trials with the strategy of ‘drug repurposing’ against COVID-19, analyzed them as per their chemical structure and possible biological targets. Results It was noticed that some of the early repurposed drugs like chloroquine, hydroxychloroquine, ACE inhibitors and ARBs, did not succeed and remained controversial. While many of the antiviral drugs like remdesivir, favipiravir, lopinavir, ritonavir, oseltamivir could be taken for the clinical trials in various countries, remdesivir could succeed to a great extent as compared to other drugs. WHO has come up with an initiative known as multi-country ‘Solidarity Trial’ for developing a potential drug or therapy against COVID-19. However, the most preferred drugs used for repurposing like hydroxychloroquine and remdesivir have not shown predictable results in solidarity trials. Conclusions The analyses of several ongoing and partially concluded clinical trials suggest that drug repurposing can be one of the major strategies for the treatment of COVID-19. Further, guidelines framed by the WHO through Infection Prevention and Control for monitoring the widespread of this COVID-19 across the world is another aggressive attempt finding the solution for the treatment for COVID-19.
Article
Full-text available
Chronic hepatitis C virus (HCV) infection in patients with cirrhosis is difficult to treat. In patients with chronic hepatitis C but without cirrhosis, once-weekly administration of interferon modified by the attachment of a 40-kd branched-chain polyethylene glycol moiety (peginterferon alfa-2a) is more efficacious than a regimen of unmodified interferon. We examined the efficacy and safety of peginterferon alfa-2a in patients with HCV-related cirrhosis or bridging fibrosis. We randomly assigned 271 patients with cirrhosis or bridging fibrosis to receive subcutaneous treatment with 3 million units of interferon alfa-2a three times weekly (88 patients), 90 microg of peginterferon alfa-2a once weekly (96), or 180 microg of peginterferon alfa-2a once weekly (87). Treatment lasted 48 weeks and was followed by a 24-week follow-up period. We assessed efficacy by measuring HCV RNA and alanine aminotransferase and by evaluating liver-biopsy specimens. A histologic response was defined as a decrease of at least 2 points on the 22-point Histological Activity Index. In an intention-to-treat analysis, HCV RNA was undetectable at week 72 in 8 percent, 15 percent, and 30 percent of the patients treated with interferon alfa-2a and with 90 microg and 180 microg of peginterferon alfa-2a, respectively (P=0.001 for the comparison between 180 microg of peginterferon alfa-2a and interferon alfa-2a). At week 72, alanine aminotransferase concentrations had normalized in 15 percent, 20 percent, and 34 percent of patients, respectively (P=0.004 for the comparison between 180 microg of peginterferon alfa-2a and interferon alfa-2a). In the subgroup of 184 patients with paired liver-biopsy specimens, the rates of histologic response at week 72 were 31 percent, 44 percent, and 54 percent, respectively (P=0.02 for the comparison between 180 microg of peginterferon alfa-2a and interferon alfa-2a). All three treatments were similarly tolerated. In patients with chronic hepatitis C and cirrhosis or bridging fibrosis, 180 microg of peginterferon alfa-2a administered once weekly is significantly more effective than 3 million units of standard interferon alfa-2a administered three times weekly.
Article
Full-text available
The hepatic stellate cell (HSC) is now well established as the key cellular element involved in the development of hepatic fibrosis and because of this there is considerable interest in establishing the molecular events that trigger and perpetuate HSC activation. HSC activation at the level of gene transcription requires the coordinated activity of several key transcriptional regulators of the HSC genome. The considerable advances that have been made in the past five years into the mechanisms by which specific families of transcription factors regulate the profibrogenic characteristics of the activated HSC are reviewed.
Article
Full-text available
Treatment with peginterferon alfa-2a alone produces significantly higher sustained virologic responses than treatment with interferon alfa-2a alone in patients with chronic hepatitis C virus (HCV) infection. We compared the efficacy and safety of peginterferon alfa-2a plus ribavirin, interferon alfa-2b plus ribavirin, and peginterferon alfa-2a alone in the initial treatment of chronic hepatitis C. A total of 1121 patients were randomly assigned to treatment and received at least one dose of study medication, consisting of 180 microg of peginterferon alfa-2a once weekly plus daily ribavirin (1000 or 1200 mg, depending on body weight), weekly peginterferon alfa-2a plus daily placebo, or 3 million units of interferon alfa-2b thrice weekly plus daily ribavirin for 48 weeks. A significantly higher proportion of patients who received peginterferon alfa-2a plus ribavirin had a sustained virologic response (defined as the absence of detectable HCV RNA 24 weeks after cessation of therapy) than of patients who received interferon alfa-2b plus ribavirin (56 percent vs. 44 percent, P<0.001) or peginterferon alfa-2a alone (56 percent vs. 29 percent, P<0.001). The proportions of patients with HCV genotype 1 who had sustained virologic responses were 46 percent, 36 percent, and 21 percent, respectively, for the three regimens. Among patients with HCV genotype 1 and high base-line levels of HCV RNA, the proportions of those with sustained virologic responses were 41 percent, 33 percent, and 13 percent, respectively. The overall safety profiles of the three treatment regimens were similar; the incidence of influenza-like symptoms and depression was lower in the groups receiving peginterferon alfa-2a than in the group receiving interferon alfa-2b plus ribavirin. In patients with chronic hepatitis C, once-weekly peginterferon alfa-2a plus ribavirin was tolerated as well as interferon alfa-2b plus ribavirin and produced significant improvements in the rate of sustained virologic response, as compared with interferon alfa-2b plus ribavirin or peginterferon alfa-2a alone.
Article
Chronic hepatitis C patients (472 patients) were treated with consensus interferon (CIFN) or interferon (IFN) alfa-2b for 6 months in a large multicenter trial. Efficacy was assessed by clearance of hepatitis C virus (HCV) RNA using reverse transcription polymerase chain reaction (RT-PCR) (<100 copies/mL), normalization of serum alanine aminotransferase (ALT), and histological improvement. The purpose of these analyses was to compare these efficacy parameters in nonfibrotics, fibrotics, and cirrhotics. Patients with chronic HCV and cirrhosis showed the same benefit from IFN treatment as noncirrhotic patients when efficacy was assessed by clearance of serum HCV RNA or by histological benefit. Sustained HCV RNA response rates were similar when measured among nonfibrotic (11%), fibrotic (13%), and cirrhotic (11%) patients. Improvement in histologic activity index (HAI) scores was noted among all 3 groups. Cirrhotic patients had a lower sustained ALT response rate (12%) than did nonfibrotic patients (23%). Ninety percent of nonfibrotics, but only 71% of fibrotics and 67% of cirrhotics, who sustained a virological response normalized their ALT. This suggests that cirrhotic patients may clear the hepatitis C virus without normalization of ALT levels. The pattern of both HCV RNA clearance over time and ALT decrease was similar among nonfibrotics, fibrotics, and cirrhotics. Tolerability to IFN therapy was similar among the 3 groups except that more cirrhotics required dose reduction because of thrombocytopenia. In patients with cirrhosis, ALT levels may be a less appropriate endpoint in the measurement of response to therapy. We conclude that liver cirrhosis should not be a reason for excluding patients from therapy because both cirrhotic and fibrotic HCV patients benefit from IFN therapy not only by clearance of virus but by improvements in liver histology.
Article
Exposure of rats to peroxisome proliferators induces members of the cytochrome P450 4A (CYP4A) family. In rats, the CYP4A family consists of four related genes, CYP4A1, CYP4A2, CYP4A3, and CYP4A8. We are specifically interested in examining CYP4A1, CYP4A2, and CYP4A3, each of which is expressed in a tissue-dependent and sex-dependent manner. While CYP4A1 is sufficiently different from the other two members to enable relatively easy specific quantitation, the close similarity between CYP4A2 and CYP4A3 makes quantitative discrimination difficult. We have combined a fluorescent real-time PCR assay (TaqMan) with the sequence-specific mismatch amplification mutation assay (MAMA) to allow us to carry out specific quantitation of all three members of this family. The assay is designed such that a single fluorescent TaqMan(R) probe binds to all three gene products, while specificity is conferred by sequence-specific primers. This specific MAMA technique takes advantage of the ability of Taq polymerase to distinguish between the two cDNAs based on mismatches at the 3' end of a PCR primer. In the 84-base PCR product used for this assay, there is only a single-base difference between CYP4A2 and CYP4A3. Despite this similarity, there is at least a 1000-fold discrimination between the two sequences, using CYP4A2 or CYP4A3 specific standards. Analysis of rat liver RNA from both sexes demonstrates that this discrimination is also achieved in complex RNA mixtures. This technique should be broadly applicable to other areas of research such as allelic discrimination, detecting mutational hotspots in tumors, and discrimination among closely related members of other gene families.
Article
A sustained virological response (SVR) rate of 41% has been achieved with interferon alfa-2b plus ribavirin therapy of chronic hepatitis C. In this randomised trial, peginterferon alfa-2b plus ribavirin was compared with interferon alfa-2b plus ribavirin. 1530 patients with chronic hepatitis C were assigned interferon alfa-2b (3 MU subcutaneously three times per week) plus ribavirin 1000-1200 mg/day orally, peginterferon alfa-2b 1.5 microg/kg each week plus 800 mg/day ribavirin, or peginterferon alfa-2b 1.5 microg/kg per week for 4 weeks then 0.5 microg/kg per week plus ribavirin 1000-1200 mg/day for 48 weeks. The primary endpoint was the SVR rate (undetectable hepatitis C virus [HCV] RNA in serum at 24-week follow-up). Analyses were based on patients who received at least one dose of study medication. The SVR rate was significantly higher (p=0.01 for both comparisons) in the higher-dose peginterferon group (274/511 [54%]) than in the lower-dose peginterferon (244/514 [47%]) or interferon (235/505 [47%]) groups. Among patients with HCV genotype 1 infection, the corresponding SVR rates were 42% (145/348), 34% (118/349), and 33% (114/343). The rate for patients with genotype 2 and 3 infections was about 80% for all treatment groups. Secondary analyses identified bodyweight as an important predictor of SVR, prompting comparison of the interferon regimens after adjusting ribavirin for bodyweight (mg/kg). Side-effect profiles were similar between the treatment groups. In patients with chronic hepatitis C, the most effective therapy is the combination of peginterferon alfa-2b 1.5 microg/kg per week plus ribavirin. The benefit is mostly achieved in patients with HCV genotype 1 infections.
Article
Modulation of gene expression through altered transcription regulates stellate cell behavior in normal liver and following hepatic injury. Transcription factors are generally classified according to conserved motifs within either the activation- or DNA- binding domains of the molecules. Transcriptional activity in stellate cells represents a delicate fine tuning of multiple inputs. Activities of these transcription factors are modified by their intracellular localization, rate and pathway of degradation, oligomerization, and interactions with heterologous factors and chromatin, as well as by posttranslational modifications, including phosphorylation, glycosylation, and acetylation. General paradigms of transcriptional control are increasingly being validated in hepatic stellate cells, particularly involving the transcription factors CCAAT/enhancer-binding proteins, c-myb, CREB, nuclear factor kappaB, peroxisome proliferator-activated receptor, and Kruppel-like zinc finger factors. Although there are no simple rules that govern mechanisms of transcriptional regulation in stellate cells, continued advances will yield new insights into their role in normal liver homeostasis and in the response to injury.