ArticlePDF AvailableLiterature Review

Cytokine-Mediated Immunopathogenesis of Hepatitis B Virus Infections

Authors:

Abstract and Figures

Hepatitis B virus (HBV) infection is a worldwide health problem, with approximately one third of populations have been infected, among which 3-5 % of adults and more than 90 % of children developed to chronic HBV infection. Host immune factors play essential roles in the outcome of HBV infection. Thus, ineffective immune response against HBV may result in persistent virus replications and liver necroinflammations, then lead to chronic HBV infection, liver cirrhosis, and even hepatocellular carcinoma. Cytokine balance was shown to be an important immune characteristic in the development and progression of hepatitis B, as well as in an effective antiviral immunity. Large numbers of cytokines are not only involved in the initiation and regulation of immune responses but also contributing directly or indirectly to the inhibition of virus replication. Besides, cytokines initiate downstream signaling pathway activities by binding to specific receptors expressed on the target cells and play important roles in the responses against viral infections and, therefore, might affect susceptibility to HBV and/or the natural course of the infection. Since cytokines are the primary causes of inflammation and mediates liver injury after HBV infection, we have discussed recent advances on the roles of various cytokines [including T helper type 1 cells (Th1), Th2, Th17, regulatory T cells (Treg)-related cytokines] in different phases of HBV infection and cytokine-related mechanisms for impaired viral control and liver damage during HBV infection. We then focus on experimental therapeutic applications of cytokines to gain a better understanding of this newly emerging aspect of disease pathogenesis.
Content may be subject to copyright.
Cytokine-Mediated Immunopathogenesis of Hepatitis B
Virus Infections
Xuefen Li &Xia Liu &Li Tian &Yu Chen
Published online: 6 December 2014
#Springer Science+Business Media New York 2014
Abstract Hepatitis B virus (HBV) infection is a world-
wide health problem, with approximately one third of
populations have been infected, among which 35%of
adults and more than 90 % of children developed to
chronic HBV infection. Host immune factors play essen-
tial roles in the outcome of HBV infection. Thus, ineffec-
tive immune response against HBV may result in persis-
tent virus replications and liver necroinflammations, then
lead to chronic HBV infection, liver cirrhosis, and even
hepatocellular carcinoma. Cytokine balance was shown to
be an important immune characteristic in the development
and progression of hepatitis B, as well as in an effective
antiviral immunity. Large numbers of cytokines are not
only involved in the initiation and regulation of immune
responses but also contributing directly or indirectly to the
inhibition of virus replication. Besides, cytokines initiate
downstream signaling pathway activities by binding to
specific receptors expressed on the target cells and play
important roles in the responses against viral infections
and, therefore, might affect susceptibility to HBV and/or
the natural course of the infection. Since cytokines are the
primary causes of inflammation and mediates liver injury
after HBV infection, we have discussed recent advances
on the roles of various cytokines [including T helper type
1 cells (Th1), Th2, Th17, regulatory T cells (Treg)-related
cytokines] in different phases of HBV infection and
cytokine-related mechanisms for impaired viral control
and liver damage during HBV infection. We then focus
on experimental therapeutic applications of cytokines to
gain a better understanding of this newly emerging aspect
of disease pathogenesis.
Keywords Hepatitis B virus .Cytokine .
Immunopathogenesis .Immunotherapy
Introduction
Hepatitis B virus (HBV) is the most prevalent virus that has
been implicated in causing acute and chronic hepatitis.
Immune-mediated mechanisms have been found to play im-
portant roles in various clinical presentations of HBV infec-
tion. In humans, the rate of chronicity is inversely proportional
to age, such as only 35 % of adults infected by HBV
persisting for more than 6 months can lead to chronic infec-
tion, whereas >90 % of children infected with HBV tend to
develop chronic infection [1,2]. Meanwhile, the risks of
developing liver cirrhosis and hepatocellular carcinoma are
greatly enhanced in patients with chronic HBV infection [2,
3]. The differences among hosts immune response can be one
of the reasons for the development and progression of hepa-
titis B [4,5]. Recent years, emerging evidences have sug-
gested that cytokines and regulatory molecules should be
regarded as fundamental mediators in determining the hosts
innate and adaptive immune responses to HBV and viral
clearance, and, therefore, the level of cytokine expression
may play a key role in disease outcome and effective antiviral
immunity (Table 1)[57,58].
Cytokines represent a large family of molecules, including
T helper type 1 cells (Th1)-associated cytokines [e.g., inter-
leukin (IL)-2, interferon (IFN)-γ], which have a functional
contribution to cellular immune responses; T helper type 2
cells (Th2)-associated cytokines(e.g., IL-4, IL-6, IL-10),
which have roles in the humoral immune responses; regulato-
ry T cells (Treg)-associated cytokines [e.g., tumor growth
X. Li :X. Liu :L. Tian :Y. Chen (*)
State Key Laboratory for Diagnosis and Treatment of Infectious
Diseases, First Affiliated Hospital, School of Medicine, Zhejiang
University, 79 Qingchun Road, Hangzhou 310003, Peoples
Republic of China
e-mail: zychenyu@126.com
Clinic Rev Allerg Immunol (2016) 50:4154
DOI 10.1007/s12016-014-8465-4
factor-beta (TGF-β), IL-10], which have been associated with
immunomodulation and immunosuppression; and T helper
type 17 cells (Th17)-associated cytokines (e.g., IL-17, IL-22,
IL-23), which play critical roles in mediating inflammation
[43]. Cytokines initiate downstream signaling pathway activ-
ities by binding to specific receptors expressed on the target
cells and play important roles in the responses against viral
infections and, therefore, might affect susceptibility to HBV
and/or the natural course of the infection [59].
Although many immune cells are able to secrete cytokines,
current information suggests that cytokines are mainly re-
leased by T lymphocytes and macrophages, which play
Tabl e 1 Profiles of cytokines
Cytokine Cell sources Primary effects Effects in HBV infection References
IFN
family
T cells; NK cells Activates monocytes and endothelial
cells;
increases class
I and II MHC molecules
Virus elimination via enhancing monocyte
macrophages, NK, and CD8
+
T cells; inhibits
HBV replication; contributes to HBeAg
seroconversion
[610]
TNF
family
T cells;
macrophages
Acutely infected phase;
inflammation; fever
Liver inflammation and hepatocytes injury; mediates
viral hepatitis complications
[1113]
IL-2 T cells; NK
cells
T, B cells growth and monocytes
activation
HBV clearance via enhancing CTL and NK cells;
promotes Treg function and causes serious side
effects by administrating high doses
[4,14,15]
IL-12 B cells;
macrophages;
NK; dendritic
cells
Differentiation of naive T cells into a
Th1 cells
Eliminates HBV via promoting and restoring CTL,
NK cells; alleviates liver damage; prompts
regeneration of hepatocytes; promotes HBeAg
seroconversion during acute HBV infection
[1620]
IL-18 Macrophages;
Kupffer cells;
NK; T cells
CTL and NK cells activation;
balance regulation
of Th1/Th2 cells; promotes
cytokines secretion
On one hand, IL-18 associating with hepatic
deterioration; on the other hand, IL-18 improves
liver damage via inducing Th1 cells and enhancing
CTL
[2123]
IL-4 T cells Differentiation of naive T cells into
Th2 cells;
regulation of humoral immunity;
macrophages
activation
Suppresses Th1-type response, maintains persistent
HBV replication and promotes immune tolerance;
paradoxically, IL-4 mediates antiviral response via
enhancing NK and T cells secreting IFN and IL-12
[2426]
IL-6 T cells;
macrophages
B cells growth and differentiation;
promoting acute
phase protein synthesis for acute
phase reactant;
inflammation
Induction of cirrhosis and HCC; eliminating HBV via
inducing humoral and cellular immune response;
resistant to lamivudine during CHB
[2730]
TGF-βMacrophages; T
cells
Induction of Treg cells and increase
Th17 cells
Inhibits HBVreplication by reducing certain transcription factor, while
contributing to HBV persistence by reducing NK functions and
mediates complications
[3134]
IL-10 T cells (Th2) Inhibition of proinflammation
cytokines; regulation
of humoral immunity; termination
of
inflammation
Sustains HBV replication and initiates chronic HBV
infection; directs fibrogenesis via inhibition of
IFN-γsecretion
[3537]
IL-17 T cells (Th17) Mediation of proinflammation
response via inducing IL-6, IL-8,
and TGF-β
Association with hepatitis, cirrhosis and HCC via combination with
proinflammation cytokines; antiviral effects via inhibiting HBV
replication; promotes stellate cells and fibroblast proliferation
[3842]
IL-23 Dendritic cell;
macrophages
Differentiation of naive T cells into a
Th17 cells;
APCs and CD4
+
T cells activation
Regulation of HBV-mediated hepatitis via IL-23/IL-17 axis [4345]
IL-22 T cells (Th17,
Th22); NK
and NKT
cells
Against bacterial pathogens in
mucosal immune
system
Prevents liver damage in ConA-induced hepatitis,
while maybe contributing to reverse effects via
recruiting inflammatory cells
[4651]
IL-21 T cells (Th17);
Tfh cells;
NKT cells
Bcells,CD4
+
T and CD8
+
T cells activation,
proliferation and survival;
induces Th17 cells differentiation;
inflammation and
autoimmune disease
Correlated with end-stage liver disease; age-
dependent response to HBV clearance
[5256]
42 Clinic Rev Allerg Immunol (2016) 50:4154
fundamental roles in immune response and are important in
pathophysiological processes, including cellular immunity,
humoral immunity, cell proliferation and differentiation, and
damage repair (Tables 1and 2)[43,64]. With further aware-
ness of the characteristics of Th1, Th2, Th3, and Th17 cells,
understanding the functional features of cytokines secreted by
these lymphocyte subgroups on immune regulation and ef-
fects is important in identifying the pathogenesis of clinical
immune-mediated diseases.
This review describes the current knowledge about how
these cytokines, such as IFN-γ, tumor necrosis factor-alpha
(TNF-α), IL-2, IL-4, TGF-β, IL-10, IL-17, and IL-22, influ-
ence the variable outcome of HBV infection and discusses
their immune signatures associated with clinical recovery and/
or persistent infection.
Th1-Associated Cytokines and HBV Infection
Th1 cells are differentiated from native CD4
+
T cells (Th0
cells) in the presence of IL-12 and IL-18 and mediate cellular
immune responses by producing anti-inflammatory cytokines
such as IFN-γ, IL-2, and TNF-α[6,16,43]. Th1-associated
cytokines are well known for playing a central role in HBV
clearance of both acute and persistent infection and also
contribute to immunopathology and influence the pathogene-
sis of liver disease [6,59].
Interferon
IFN has a pivotal role in the resolution of an acute hepatitis B
episode and may be the main mediator in noncytolytic control
of HBV infection [7]. Increased expression of major histo-
compatibility complex (MHC) molecules on the cell surface is
the main antiviral mechanism of IFN, contributing to antigen
presentation to CD8
+
T cells and leading to dissolution of
target cells. Meanwhile, it can enhance phagocytosis in NK
cells and monocytemacrophages to target cells infected with
the virus, as well as inhibiting viral replication in newly
infected cells, which is conducive to virus elimination [7,8].
Virus replication often results in rapid induction of IFN by
the infected cell [65]. A robust activation of IFN-γproduction
by innate and adaptive immune cells (e.g., NK and NKT cells,
HBcAg specific CD4
+
and CD8
+
Tcells)in self-resolving
acute HBV infection is characterized with significant inhibi-
tion of HBV replication [6,8]. Several HBV-related studies
have characterized that IFNs can inhibit the HBV replication
cycle both transcriptionally and post-transcriptionally [9]and
are capable of preventing the formation of new HBV capsids,
leading to the destabilization of existing capsids [10,66,67].
Tabl e 2 Clinical characteristics and cytokine responses in the different stages of HBV infection
Stages of HBV
infection
Immune responses
mediated by
cytokines
Clinical manifestation References
Markers of
the virus
Serum
biochemical
parameters,
e.g., ALT
Liver
histopathology
Treatment
Occult HBV infection IL-10,TGF-β,
IL-17, IL-21
HBsAg ()
HBeAg ()
Anti-HBe (+/)
HBV-DNA (+)
Normalization No liver
damage
No
therapy
[1,4,6062]
Inactive carrier IL-10, TGF-βHBsAg (+)
HBeAg ()
Anti-HBe (+/)
HBV-DNA
(<2000 IU/ml)
Normalization Low-grade
inflammation
No
therapy
[1,4,60,61]
Acute self-limit
HBV infection
IL-12,IL-18, IFN-α,
IL-21
IFN-γ,IL-22
IL-6, TNF-α
IL-10,
HBsAg (+)
HBeAg (+)
Anti-HBe (+/)
HBV-DNA (+)
Minimal or
temple Liver
damage
No
therapy
[4,5,51,60]
Active chronic
HBV infection
IL-12↑↑, IL-18↑↑,IFN-α,
IL-10, IL-17, IL-6
HBsAg (+)
HBeAg (+/)
Anti-HBe (+/)
HBV-DNA (+)
↑↑ Increased liver
damage
Antiviral
therapy
[4,20,60,61,63]
HBV hepatitis B virus, IL interleukin, TGF-βtumor growth factor-beta, IFN-αinterferon-alpha, HBsAg hepatitis B surface antigen, HBeAg hepatitis B e
antigen, Anti-HBe antibody to HBeAg, ALT alanine aminotransferase, () negtive, (+) positive, (+/) positive or negative, decline, elevation, ↑↑
significant elevation
Clinic Rev Allerg Immunol (2016) 50:4154 43
However, the lack of large IFN-γand TNF-αproduction
by weaker virus-specific T cells has been observed in chronic
HBV patients, who may fail to develop an efficient antiviral-
specific immune response, contributing to virus persistence
and chronic liver immunopathology [8,58]. Induction of
serum IFN-γlevels on treatment in chronic hepatitis B
(CHB) patients can lead to virological control and HBeAg
seroconversion [68].
Tumor Necrosis Factor-Alpha
TNF-αis considered to be a key cytokine involved in the
immune pathogenesis of HBV infection [11]. TNF-αnot only
participates in the hosts anti-HBV immune response but also
mediates liver inflammation and hepatocytesinjury after
HBV infection. For instance, HBV infection leads to elevated
levels of TNF-α, which is relevant to cirrhosis and hepatic
encephalopathy (HE) [12,13].
Studies have shown that serum levels of TNF-αand its
receptor are significantly increased in HBV-infected patients
[57,69]. Genetic analysis revealed an association between the
polymorphism in the promoter region of TNF-αlocated at
308G/A and HBV disease progression [70,71], while an-
other study proposed that TNF-αpromoter polymorphisms (at
positions 238A, 308A, 857T, 863A, and 1031C) are
important host genetic factors that may determine the clinical
outcome of HBV infection [72]. These data may indicate the
role of TNF-αin the pathogenesis of hepatitis B as well as its
complications.
Interleukin-2
IL-2, also known as T-cell growth factor, is primarily activated
by T cells and partly produced by NK cells and transformed B
cells, which exerts biological effects through combination to
cell surface receptor IL-2R. IL-2 can promote proliferation
and differentiation of T cells and B cells, activate monocytes/
macrophages, and enhance the killing activity of cytotoxic T
lymphocytes (CTL) and NK cells and thus is a vital factor in
the regulation of the immune response [72].
IL-2 production by HBV-specific CD4
+
T cells plays a
major role in the efficient development of effector cytotoxic
CD8
+
T cells and contributes to HBV clearance in human
HBV infection by preventing the virus from stimulating T
cells, B cells, and macrophages to express IL-2R simulta-
neously. T cells with an expression of IL-2R are direct effector
cells that cause liver damage. Meanwhile, the decline of IL-2
weakens the antiviral ability of host and promotes viral repli-
cation, both of which form a vicious circle [14].
Recent results indicate that IL-2 is a potential activator of
memory and effector T cells and can also restore effects and
functions of tolerated T cells to antigens. Thus, many re-
searchers have aimed to focus on whether IL-2 can be applied
in the treatment of hepatitis B. However, high doses of sys-
temic administration of IL-2 not only cause serious side ef-
fects but also promote the function of Treg cells and increase
the activation threshold of antigen-specific T cells [15,73].
Therefore, whether IL-2 can become a therapy target needs
further investigation.
Interleukin-12
IL-12 is a type of multifunctional cytokine mainly secreted by
B cells, monocytes/macrophages, and dendritic cells, which
induce immune responses against viral infections, especially
for HBV. IL-12 can induce Th0 cells to differentiate into Th1
cells, which produce type I cytokines (such as IL-2 and
IFN-γ) and inhibit Th2 cells so that IL-4 and IL-6 cannot be
synthesized. In addition, IL-12 can also cooperate with IL-2 to
promote proliferation and differentiation of cells including
CTL and NK, and help CTL cells, which help eliminate
virus-infected hepatocytes [16].
Studies indicate that, in patients with persistent HBV in-
fection, a decrease in IL-12 can prompt the balance of Th1/
Th2 to tilt forward Th2 and cause noncytotoxic T lymphocyte
response against virus clearance, resulting in chronic HBV
infection [17]. IL-12 can restore consumption of CD8
+
Tcells
and substantially reduce HBV-specific CD8
+
T cells exhaus-
tion. Additionally, it was able to compete against TNF, alle-
viate liver damage from the virus, prompt regeneration of
hepatocytes, improve phagocytosis of Kupffer cells, and pre-
vent further endotoxin damage to liver cells [18,74].
Clinical studies also revealed that IL-12 plays important
roles in HBV infection; patients with long-term forms of
hepatitis B are unable to produce a sufficient amount of this
cytokine [19]. In acute HBV infection, a high concentration of
IL-12 usually occurs, which may be one of the factors that
promote spontaneous negative conversion of HBeAg. During
the process of antiviral therapy, a low serum level of IL-12
suggests the bodys undesirable reaction to antiviral drugs [20,
35]. Decline of IL-12 in chronic hepatitis B patients may be
closely related to maturation and dysfunction of mDC [17].
Interleukin-18
IL-18 is a type of newly discovered multifunctional cytokine,
whichismainlysecretedbyactivatedmonocytemacro-
phages, Kupffer cells, NK cells, T cells, etc., affecting the
balance regulation of immune responses. IL-18 can initiate a
nonspecific immune response through NK cells, participate in
the balance regulation of Th1/Th2 cells, increase specific
immune response of cytotoxic T lymphocytes (CTL), as well
as enhance other cytokines secretion, which is significant in
immune regulation after pathogen infection [21].
After HBV infection, NK cells are nonspecifically stimu-
lated by IL-18 to secrete IFN-γ, followed by the production of
44 Clinic Rev Allerg Immunol (2016) 50:4154
IFN-αand IFN-β; these cytokines can further lead to hepato-
cyte dysfunction while inhibiting viral replication. Studies
indicate that the IL-18 level increases with the concentration
enrichment of HBV DNA. On the one hand, with the syner-
gistic effect of IL-12, IL-18 can induce Th1 cells to produce
large amounts of IFN-γ, thus exerting antiviral function;
however, in an immune microenvironment with insufficient
IL-12, IL-18 can only activate Th2 immune response; thus,
the ability of viral abolition declines, and it finally turns to
chronic infection [22]. On the other hand, the abundant IFN-γ
induced by IL-18 can stimulate CD8
+
T cells to express Fas
and upregulate the expression of soluble intercellular adhesion
molecules (SICAM-1) in liver cells, enhancing the cytotoxic-
ity of CTL. Meanwhile, SICAM-1 mediates effector cells to
attack hepatocytes and thereby aggravating hepatocytesdam-
age in the inflammatory sites of the HBV infection [23,75].
Altogether, IL-18 is a double-edged sword in the pathogenesis
of viral hepatitis; high expression of IL-18 is in favor of virus
clearance but can also lead to sustained chronic liver inflam-
mation. To clarify the role of IL-18 in the pathogenesis of
hepatitis B and hepatocyte injury will provide a new approach
for the prevention and treatment of this refractory disease.
Th2-Associated Cytokines and HBV Infection
Th2 cells are related to proliferation and differentiation of B
cells, as well as antibody generation. Th2 cells mediate the
humoral immune response mainly by secreting cytokines such
as IL-4, IL-6, etc. Th1 type cellular response can be restrained
by Th2 cells and cause a reduced antiviral ability in the host. A
disorder of Th1/Th2 balance is closely associated with the
outcome of HBV infection: When Th1 type response domi-
nates, the body presents self-limited or acute fulminant hepa-
titis; however, when the response of Th2 type dominates, the
body is more likely to turn to chronic hepatitis. Therefore, a
disorder of Th1/Th2 balance is the main factor that causes
chronic HBV infection [76,77].
Interleukin-4
IL-4, mainly secreted by activated Th2 cells, holds the bio-
logical effects of stimulating proliferation and differentiation
of B cells to produce antibodies, promoting CD4
+
Tcellsto
differentiate into Th2 cells, and improving activity of macro-
phages, which is of great significance in the regulation of
humoral and adaptive immunity [24].
An elevated IL-4 level in the peripheral blood of HBV-
infected patients can suppress the production of IFN-γand
Th1-type immune response, resulting in persistent HBV rep-
lication and promotes the formation of immunetolerance [25].
Paradoxically, it was previously reported that IL-4 can
promote T cells and NK cells to secrete IFN-γindirectly by
stimulating dendritic cells (DC) to produce IL-12 without
depending on IL-12; in addition, IL-4 might suppress the
production of HBsAg, as well as the replication of HBV and
thus mediate the hosts antiviral response [24,26]. Building on
the previous sections, it can be seen that IL-4 has dual roles in
the prolonged forms of hepatitis B and further disease
progression.
Interleukin-6
IL-6 is a key cytokine in the bodys immune regulatory
network and is secreted from cells existing in each system of
the human body. The primary biological effects consist of
regulating growth and differentiation of B cells, promoting
synthesis of acute phase protein from hepatocytes, and en-
hancing the killing effect of CTL and NK cells, leading to liver
inflammation and immune damage [27].
IL-6 plays dual roles in the pathogenesis of hepatitis B;
importantly, it can protect the liver from virus infections by
stimulating immune responses against hepatocytes. However,
IL-6 also has an important role in the induction of hepatitis,
cirrhosis, and hepatocellular carcinoma (HCC) [27]. Numer-
ous studies have illustrated that serum IL-6 expression was
significantly higher in CHB patients than that in naturally
immune subjects, which plays key roles in the elimination of
HBV with the induction of humoral and cellular immune
responses [28,29,78]. In parallel with the mentioned study,
another investigation reported that HBV infection leads to
elevated levels of IL-6 in CHB patients who were resistant
to lamivudine in comparison with patients maintaining a
normal response [30]. In addition, it has been established that
this cytokine may participate in pathological complications of
hepatitis B [79]. Based on the presented studies, it appears that
IL-6 is a favorable candidate to induce immune responses
against HBV.
Treg-Associated Cytokines and HBV Infection
Treg cells is a major T cell subset with immunosuppressive
function, which mainly produce immunosuppressive cyto-
kines such as TGF-β, IL-10, and IL-35 and inhibit the activa-
tion of Th1 or Th2 cells, downregulate the immune response,
and induce host immune tolerance, resulting in long-term
persistence of the virus [1,80]. A high viral load has been
shown to suppress CD4
+
and CD8
+
T cells in addition to the
induction of Treg cells in patients with chronic HBV infection
[81,82]. The level of peripheral Treg cells is significantly
increased in patients with chronic hepatitis B. TGF-β,IL-10,
and IL-35 secreted by Treg cells are strongly linked to liver
Clinic Rev Allerg Immunol (2016) 50:4154 45
cirrhosis and primary hepatocellular carcinoma caused by
chronic hepatitis B [31,81,83].
Tumor Growth Factor-Beta
TGF-βis a 25-kDa cytokine withthree homodimeric isoforms
in humans, including TGF-β1, TGF-β2, and TGF-β3[84].
The TGF-β1 isoform is the most widely expressed isoform of
the family and is referred to as TGF-βin many studies. A
previous investigation demonstrated that TGF-βplays a dual
role in the regulation of immune system and in the responses
against viral infections. TGF-βnot only induces immune
tolerance against HBVantigens via stimulating the differenti-
ation of Treg cells, but also, it has an important role in the
development of cirrhosis of the liver and HCC via increasing
Th17 cells [31,38,85].
Recent findings also indicate that TGF-βcan be considered
as either good or bad for hepatitis B. For example, Hong et al.
reported that TGF-βcan inhibit HBV replication by reducing
hepatocyte nuclear factor-4alpha expression, which is an im-
portant transcription factor required for HBV suppression
[32]. Supporting the role of inhibiting HBV, another investi-
gation suggested that TGF-βcan directly alter hepatocyte
function by regulating micro-RNA expression in hepatocytes
and downregulates NKG2D/DAP10 and 2B4/SAP expression
in NK cells, thus contributing to HBV persistence [33,34,86].
Therefore, the elevated levels of TGF-βcanberegardedasa
crucial step in the mechanism by which HBVescapes the host
immune system. Future studies are needed to improve the
knowledge of the putative roles and detailed mechanisms that
are affected by TGF-βin the pathogenesis of HBV-mediated
hepatitis B and its complications.
Interleukin-10
IL-10, a unique cytokine that was originally discovered in the
murine secretion of Th2 cells, attracts wide concerns from
domestic and foreign scholars because of its inhibition in the
synthesis of Th1 cytokines [87]. IL-10 owns the main biolog-
ical effects of inhibiting the production and release of proin-
flammatory cytokines, thus limiting and terminating inflam-
mation, and promoting the proliferation and differentiation of
B cells to produce antibodies [36].
Previous studies demonstrate that increased expression of
IL-10 is the primary factor that causes chronic HBV infection
[37]. IL-10 can restrain the hosts anti-HBVactivity, leading to
sustained replication and expression of HBV in vivo. An
elevated level of IL-10 is relevant to HBV serum titers and
degree of liver inflammation and also directs the processes of
fibrogenesis and further disease progression [35,37]. Howev-
er, IL-10 is able to induce the activation and proliferation of B
cells and NK cells and enhance the cytotoxicity of CD8
+
T
cells, while inhibiting the production of IFN-γselectively
without changing the cytotoxicity of NK cells. Results obtain-
ed by in vivo and in vitro studies have identified that blockage
of the IL-10 receptors could recover HBV specific CD8
+
T-
cell response, accelerate virus elimination, and suppress the
chronicity of viral infection [37,88]. Therefore, studies of the
effects of IL-10 on HBV infection need to be evaluated by
more investigations.
Th17-Associated Cytokines and HBV Infection
Th17 cells are a newly identified subset of T helper cells and
are closely related to the progression of HBV disease [39].
Research interest in these cells has indicated that patients with
chronic HBV infection were found to have significantly ele-
vated Th17 cell frequency and Th17-secreted cytokines, in-
cluding IL-17A, IL-21, and IL-22, and it was proposed that
these proinflammatory effectors may perform a vital function
in pathogenesis of prolonged hepatitis B infection.
Interleukin-17
IL-17 is considered to be the principal mediator of Th17 cell
functions. The cytokines of the IL-17 family have been re-
ported to be involved in the initiation and maintenance of
immune processes, most notably in inducing and mediating
proinflammatory responses. It has been established that IL-17
binding to the IL-17 receptor on target cells can induce the
expression and secretion of proinflammatory factors, such as
IL-6 and IL-8, which in turn promotes the inflammatory
response [40,41].
Previous studies identified that IL-17 is upregulated in
chronic HBV-mediated inflammation and could be relevant
to the development of liver cirrhosis and HCC [38,39,42]. IL-
17 exerts its antiviral effects by inducing the expression of
MxA and OAS both from messenger RNA (mRNA) levels
and protein levels, which directly inhibits HBV replication
[89]. In addition, IL-17 can also significantly stimulate mono-
cytes and DCs to express IL-17R and produce proinflamma-
tory cytokines, such as IL-1β,TNF-α, IL-6, etc., which are
important for liver damage during hepatitis B progression
[63]. Studies revealed that IL-17 directly mediated the inflam-
matory response in liver cells, and activation of the inflam-
matory response in liver tissue between the liver stromal cells
abundantly expressed TGF-β1, resulting in the conversion to
liver fibrosis in chronic hepatitis B [39].
Supporting the role of prompting stellate cell and fibroblast
proliferation, another investigation suggested that hepatic stel-
late cells also can express IL-17R, and IL-17 can directly
affect IL-17R-expressing cells and exacerbate the inflamma-
tory microenvironment of the liver [63]. Building on the
previous sections, it can be seen that IL-17 may play a critical
46 Clinic Rev Allerg Immunol (2016) 50:4154
role and exhibits a potential to exacerbate liver damage during
chronic HBV infection. Hence, the phenomenon that IL-17
preferentially increased in HBV infected patients may support
a role for IL-17 in the immunopathogenesis of chronic HBV-
mediated inflammation, but the exact mechanism of action
needs to be investigated further.
Interleukin-23
IL-23 is a recently discovered heterodimeric cytokine of an
IL-12 shared p40 subunit and a unique p19 subunit [90]. It has
been shown that IL-23 regulates a myriad of processes in the
innate and adaptive immune response, including promoting
CD4
+
T-cell proliferation and IFN-γand IL-12 production,
enhancing the antigen presentation abilities of antigen pre-
senting cells (APCs) [44]. IL-23 was also shown to efficiently
stimulate the differentiation of naive CD4
+
TcellsintoTh17
cells in the presence of HBsAg or HBcAg and is an important
mediator of the proinflammatory effects exerted by Th17 cells
in many diseases [43,45].
More recently, it has been reported that HBV-infected
patients have a positive association between IL-23 cytokine
level and Th 17 cell axis and, hence, might play an important
role in the pathogenesis of hepatitis B [43,45]. A study by
Wang et al. [45] indicated that HBsAg and HBcAg can induce
mDCs and macrophages to secrete large amounts of IL-23
protein in a dose-dependent manner in vitro. The IL-23/IL-
23R pathway was strongly activated in liver tissues from
HBV-infected patients. Moreover, IL-23 derived from mDCs
and macrophages can stimulate the capacity to produce IL-17
from CD4
+
T cells in the presence of HBV antigen [43]. These
data provide novel insights into the mechanism of HBV-
induced hepatitis through the IL-23/IL-17 axis.
Interleukin-22
IL-22 is an IL-10 cytokine family member, and the main
sources of IL-22 are Th17 [46], Th22 [47], as well as activated
NK and NKT cells [48]. IL-22 exerts its functions via binding
to a receptor complex composed of IL-10Rβand IL-22R
chains. The latter is highly expressed within the pancreas,
liver, kidney, small intestine, and colon [91,92]. Research
concerning the role of IL-22 remains controversial.
At present, most studies support a protective role for IL-22
in preventing liver damage. In T-cell-dependent hepatitis such
as concanavalin A (ConA)-induced liver injury, IL-22 mRNA
and protein expressions were both significantly elevated in
mice injected with ConA, but they were increased less exten-
sively in the carbon tetrachloride (CCl4)-induced liver injury
model [49,93]. Furthermore, overexpression of IL-22 in the
liver defends against liver injury. Moreover, another study
demonstrated that IL-22-deficient mice are highly sensitive
to ConA-mediated hepatitis and adoptive transfer of IL-22-
expressing Th17 cells into these mice protects against
hepatitis-induced liver injury [94]. Together with the evidence
that liver-specific IL-22 transgenic mice are resistant to hep-
atitis induced by ConA [50], the above studies support a
protective role of IL-22 in liver damage.
However, there are studies demonstrating the pathogenic
activity of IL-22. One group used a transgenic mouse model
of HBV replication and revealed that IL-22 neutralization
improved hepatic expression of proinflammatory genes,
resulting in much more severe liver damage [51]. Further-
more, IL-22 abolition in this transgenic mouse significantly
inhibited the recruitment of antigen-nonspecific inflammatory
cells into the liver. Collectively, these studies suggest that IL-
22 may contribute to the pathogenesis of liver disease by
promoting the migration of inflammatory cells into the liver,
and then plays a proinflammatory role in T-cell induced he-
patocytes [95]. In human studies, it was shown that the level of
serum IL-22 markedly increased in patients with acute hepa-
titis B (AHB), e.g., the IL-22 derived from CD4
+
Tcells [51].
Xiang et al. proposed that IL-22 was highly expressed in the
liver tissue of CHB patients [50,96], suggesting a protective
role in liver inflammation during CHB infection. However,
Park et al. [50] also reported upregulation in patients with
chronic HBV, but in his study, IL-22 levels were positively
correlated with liver inflammatory degree in CHB patients and
accelerated liver cancer promotion in these patients. To date,
there is no general consensus on whether IL-22 is involved in
the progression of HBV as a proinflammatory factor or as a
protective factor. It is possible that IL-22 could exert dual
functions; thus, future clinical investigations to explore the
exact role and design new therapeutic strategy against chronic
HBV infection of IL-22 are worth investigating.
Interleukin-21
Interleukin-21 (IL-21) is a pleiotropic cytokine, which is a
member of the type I cytokine family [97] and is produced by
Th17 cells, Tfh cells, and natural killer T (NKT) cells. The IL-
21 receptor is widely expressed on a variety of immune cells,
including CD8
+
T cells, B cells, NK cells, dendritic cells,
macrophages, and keratinocytes [98,99], and IL-21 signals
via IL-21R as well as the common cytokine receptor γ-chain.
Based on previous studies, IL-21 can affect CD4
+
T cells and
B cells through regulating their activation, proliferation, and
survival [52]. In addition, IL-21 can synergistically promote
the expansion of CD8
+
T cells and induce Th17 differentiation
[53,54] and, hence, is associated with many inflammatory and
autoimmune diseases [100].
Recently, several groups have explored the function of IL-
21 in HBV clearance and its contributions to the severity of
chronic hepatitis B (CHB). One group constructed an animal
model of human HBVand described a phenomenon that adult
mice have higher HBV-specific liver IL-21 production than
Clinic Rev Allerg Immunol (2016) 50:4154 47
young mice; the low level of IL-21 in younger mice leads to
lower frequencies of virus-specific CD8
+
T- and B-cell re-
sponses. Therefore, it was concluded that IL-21 plays a key
role in the age-dependent response to HBV clearance [55],
which is consistent with another group [56]. Moreover, they
also detected the levels of IL-21 transcripts in humans blood
samples from acutely infected adult patients who had previ-
ously cleared the virus; these patients were found to express
high levels of IL-21, in contrast to low levels expressed in
chronic hepatitis B patients or chronic HBV carriers [55].
With the above results, IL-21 is proved to have a pivotal role
in determining the age-dependent effectiveness of immune
responses. Another group showed that serum IL-21 levels
were higher in ACLF patients and severe CHB patients than
in moderate CHB patients and normal controls, which corre-
lated with end-stage liver disease (MELD) score, survival, and
recovery [101]. Thus, this study suggested IL-21 had a causal
role in the development of severe liver inflammation and was
associated with severity of liver disease.
Experimentally Therapeutic Applications of Cytokines
and Their Receptors
Currently, antiviral drugs, such as lamivudine, adefovir,
entecavir, tenofovir, telbivudine, and interferon (e.g.,
pegylated IFN-α2b), are the commonly used treatment of
chronic HBV infection as summarized in Table 3[3,60,
110]. Although anti-HBV nucleoside drugs mainly target the
viral polymerase activity and exhibit the early virological
response, the long-term effects of most antiviral drugs is still
unsatisfactory (having, e.g., the emergence of HBV mutants
and post-treatment relapse) [60,111]. Emerging evidence has
suggested that restoring the functions of HBV specific CTLs
is the key for effective antiviral therapy [112]. Thus, more
concerns and efforts have been drawn on immunotherapy as
novel therapeutic strategies for HBV [110].
Cytokines, important immunomodulators, serve central
roles in the initiation and regulation of immune responses
and, furthermore, might affect susceptibility to HBV and/or
the outcome of the infection.Much attention has been given to
enhancing T-cell immunity through the use of certain cyto-
kines, which have proven to be essential and necessary for T-
cell survival and function. Therefore, recombinant cytokines
that can be used as therapeutic agents for regulation of the
immune system and in responses against viral infection
followed the development of genetic engineering technology.
IFN-αwas considered the drug of choice for the treatment
of hepatitis B both at home and abroad. It has been reported to
stimulate a predominantly innate immune response, such as
the NK cell response, and increase the cytotoxicity and de-
crease IFN-γproduction of NK cells [113]. Futhermore,
IFN-αexerts their antiviral and virus clearance activities of
the host by inhibiting HBV transcription and replication by
targeting the epigenetic regulation of the covalently closed
circle DNA (cccDNA) viral minichromosome [105], blocking
the formation of an RNA-containing core particle [10]and
expediting the degradation of nucelocapsids that contain
pregenomic HBV RNA [106]. The treatment of IFN-αresults
in a progressive decline of serum HBV DNA, seroconversion
to anti-HBe, and anti-HBs status, as well as ALT normaliza-
tion for both HBeAg-positive and HBeAg-negative CHB
patients [102,107,114]. The combination of IFN-γplus
another Th1-type proinflammatory cytokine TNF-αand se-
quential therapy with cytokine and lamivudine showed a
stronger inhibition of HBV cccDNA compared with
lamivudine alone [115].
However, although with finite duration and absence of
resistance, IFN-αis associated with disadvantages such as
exorbitant cost, the need for subcutaneous injection, contrain-
dication profiles and significant side effects, together with a
relatively small percentage (2030 %) of HBV-infected pa-
tients who respond to IFN-α-based therapy [116].
Recently, IFN-α-based therapy has been largely replaced by
pegylated interferon-α(PEG IFN-α), where IFN-αis conju-
gated with polyethylene glycol molecules that prolongs its half-
life and facilitates the administration schedule; furthermore,
pegylation has reduced the immunogenicity and antigenicity
for the target protein, resulting in a decreased hypersensitivity
reaction [103]. Pegylated interferon-αenhances recovery of
memory T cells in antigen-positive chronic hepatitis B patients
by downregulating inhibitory receptors (PD-1 and CD244) and
upregulating effector molecules (CD127 and CXCR4) [104].
PEG IFN-α-2a and PEG IFN-α-2b are two forms of PEG
IFN-α, which can be used to treat CHB patients with a similar
efficacy in clinical trials. Currently, the recommended dose and
duration of PEG IFN-α-2a administration for HBeAg-positive
patients is 180 μg/week for 48 weeks [103]. As an authorized
monotherapy drug, PEG IFN-αhas finite duration and a more
durable PEG-IFN-induced HBeAg seroconversion [117]and,
hence, is appropriate for young people who are considering
pregnancy, genotype A patients with high or low levels of HBV
DNA, and genotypes B and C patients with both high levels of
ALT and low levels of HBV DNA [118]. Meanwhile, PEG
IFN-αis endowed with disadvantages, such as inferior tolera-
bility, risk of adverse events, and the need for parenteral ad-
ministration, which is contraindicated in patients with decom-
pensated HBV-related cirrhosis and patients receiving immu-
nosuppressive therapy or chemotherapy [61]. The limited effi-
cacy of IFN-α/PEG IFN-αmonotherapy prompted the treat-
ment optimization, thus providing the opportunity to combine
IFN-αand nucleos(t)ides analogues (NUC) therapy; the com-
bination therapy results indeed showed a slightly better antiviral
effect in HBeAg-positive and in HBeAg-negative patients than
IFN-αmonotherapy [119122].
48 Clinic Rev Allerg Immunol (2016) 50:4154
As a new biological agent, TNF-αinhibitors have been
used in the treatment of various autoimmune disease, includ-
ing rheumatoid arthritis (RA), ankylosing spondylitis (AS),
psoriasis, psoriatic arthritis (PsA), and Crohnsdisease[123,
124], but the safety of TNF-αinhibitors (such as infliximab)
in these patients with HBV infection is still controversial.
TNF-αis an important cytokine involved in host defense,
anti-TNF therapy may allow virus to escape immune control
and increase patientssusceptibility to virus infections. Since
the first case reports of HBV reactivation in patients with anti-
TNF-treatment in 2003 [125], more cases were reported and
concerned on the risk of HBV infection patients treated with
anti-TNF agents [126,127]. HBV reactivation during treat-
ment with TNF-αinhibitors in HBV carriers or chronically
infected HBV patients has been literally referred by several
case reports [128,129]. Concomitant pre-emptive antiviral
therapy could successfully reduce the risk of HBVreactivation
[130]. To the contrary, the safety of TNF blockers in HBV-
infected patients, especially the resolved/occult HBV patients,
has been demonstrated by other case reports [128,131].
However, antivirals are necessary for patients with both auto-
immune disease and HBsAg positive during TNF-αblockers
treatment. In addition, more data were needed for further
evaluations of the safety in HBV-infected patients with anti-
TNF therapy.
As one of the main cytokines involved in inducing cellular
responses against HBV infections, IL-12 [132] is indispens-
able for viral eradication of infected hepatocytes [133]. Al-
though the recombinant human IL-12 (rhIL-12) alone is nei-
ther effective nor without side effects [74], the combined
therapy of lamivudine plus rhIL-12 was able to increase T-
cell reactivity to HBV and the secretion of IFN-γ[108]. A
combination of IL-12 and IL-18 was found to strongly stim-
ulate IFN-γproduction by CD4
+
T cells isolated from the
peripheral blood in response to HBcAg and may contribute to
viral clearance in children with chronic hepatitis B infection
[109]. One study used human recombinant IL-12 directly and
indicated that rhIL-12 downregulates HBV copy numbers in
CHB patients [134]. Another study demonstrated that IL-12
strengthens the response of HBV-specific CD8
+
T cells within
the liver and promotes HBV-specific central memory CD8
+
T
cells and HBeAg seroconversion [18]. Therefore, IL-12 as an
inflammatory cytokine can be considered as a favorable can-
didate for treating HBV infection.
IL-10, an anti-inflammatory Th2 cytokine, is produced by
Th2 lymphocytes, activated macrophages, T regulatory and B
regulatory cells, and inhibits expression of Th1 proinflamma-
tory cytokines such as IFN-γ[135]. It was reported that the
higher level of IL-10 production may restrain the immune
function against HBV in patients with occult HBV infection
[62]. Moreover, inhibition of IL-10 in vitro has been proved to
rescue the polyfunctionality of virus-specific CD8
+
Tcells,
which were regulated via IL-10-producing B cells [37].
TGF-β, also an anti-inflammatory cytokine, is produced
together with IL-10 by Treg cells, which appear to be partially
responsible for liver immune tolerance [136]. In a CHB pa-
tient, inhibition of IL-10 with or without TGF-βwas able to
restore the cytotoxic capacity of liver NK cells to produce
IFN-γ, thereby enhancing their noncytolytic antiviral capacity
[137]. TGF-β1 treatment was able to effectively suppress
Tabl e 3 Effectiveness of antiviral agents for treatment of chronic hepatitis B
Activity Target Action Effect Outcome References
Antiviral: such as
Lamivudine,
Adefovir,
Entecavir,
Telbivudine,
Ten ofo vir,
pegylated IFN-α
HBV Inhibition of formation of
infectious virus;
induction of antiviral
response in cells;
regulation of HBV
cccDNA
Decrease of virus DNA and
correlative antigens (HBsAg,
HBcAg, HBeAg); improved
HBeAg seroconversion;
histological and ALT
improvement and normalization;
restored function of HBV-
specific T cells
Safe and effective in both
HBeAg-positive and negative
chronic hepatitis B; especially,
lamivudine therapy
administrated as
decompensated cirrhosis can
prolong survival and delay or
even make liver
transplantation dispensable;
combination therapy are often
used for mutant HBV strains
[2,3,60,
61,
102104]
Immune modulatory:
such as IFN-αand
its pegylated
formulations: Peg-
IFN α-2a, Peg-IFN
α-2b, rhIL-12
Hosts
innate
and
acquired
Immunity
Directly activates and
enhances NK cells
effect; increases CTL
and CD4 T cells
response
Reduction of serum HBV DNA;
increased loss of HBeAg and
HBsAg; achieved HBeAg
seroconversion; histological
improvement; normalization of
ALT; increased IFN-γproduction
and IFN-γrelated cytokines and
immune cells against virus
HBV infection effectively under
control by regulating robust
immune response
[18,103,
105109]
Clinic Rev Allerg Immunol (2016) 50:4154 49
HBV replication and the levels of viral transcripts, core pro-
tein, and nucleocapsid [32,138]; hence, TGF-β-based immu-
notherapy needs further exploration to achieve an optimal
therapeutic regimen for HBV patients.
Previous data have proposed that injection of granulocyte-
macrophage colony-stimulating factor (GM-CSF), known to
be a good adjuvant for vaccination, before the vaccine might
promote a stronger immune response in humans [139]; recom-
binant human GM-CSF significantly reduced hepatitis B virus
DNA levels [140]. Subsequently, a pilot study demonstrated
that a combined therapy of GM-CSF with a hepatitis B vac-
cine inhibited HBV replication in carrier children [141]. It was
reported that when the HBV-S gene is fused to the GM-CSF
gene, the immune responses induced by HBV-DNA plasmids
both in normal and HBV-transgenic mice can be strengthened,
and HBV-DNA plasmids fused with GM-CSF may be used
for prevention and treatment [142], but meanwhile, it also
showed that GM-CSF was not able to improve the antibody
(Ab) titer or the development of protective immunity by the
HBV vaccination in healthy adult nonresponders [142]. Re-
cently, another study revealed that GM-CSF failed to improve
responses to the booster HBV vaccination in HIV-infected
individuals [143]. Thus, whether GM-CSF is an optimal vac-
cination strategy needs to be evaluated.
Conclusion and Future Prospects
HBV is the most common cause of acute and chronic liver
disease worldwide [144]. Cytokines secreted by immune cells
are crucial in cell mediated and humoral immune responses as
well as in antiviral activity, viral clearance, apoptosis, and
fibrogenesis [109,145]. When affected by HBV, multiple
cytokines and immune cells interact with one another and
form a complex network of immune response, thus prompting
the occurrence and development of hepatitis B (Fig. 1). There
have been several attempts to develop immunotherapy, such
as cytokine therapy, for the control of chronic HBV infection,
and some results are promising. The LCMV mouse model has
showed the essentiality of IL-2 administration timing and
differentiation status in designing IL-2 therapy, indicating a
therapeutic function for IL-2 [146]; TGF-β, which has been
discussed for HBV immunotherapy, is produced by HBV-
specific CD8
+
T cells and contributes to CD8
+
T-cell restora-
tion [147150]. As a main regulative cytokine of cellular
immunity against viral infection, IL-10 possesses inhibitory
effects on cell-mediated immune responses [151]; thus, its role
in HBV treatment is under exploration. In addition, in a mouse
model of human hepatitis B, IL-21 functions in the therapeutic
augmentation of immune responses to HBV [55]. As another
Th17-secreted cytokine, IL-22 was widely studied regarding
its role in hepatitis. The dual function of IL-22 with both
proinflammatory and protective roles in liver inflammation
and patients with hepatitis B [43] reveals the necessity for
future clinical investigations. Cytokines not only play a key
role in the clearance of HBV [62] but are also indispensable
for NK cells and cytotoxic T cells to balance their function in
immune response [152,153]. Therefore, monitoring the ex-
pression of these cytokines may directly reflect the patholog-
ical condition for liver, which has instructive functions for the
treatment of hepatitis B. Collectively, insight into cytokine
function during the occurrence and development of hepatitis B
is valuable for its prevention and treatment.
Acknowledgments This paper was supported in part by a grant from
the National Natural Science Foundation of China (No. 81171565),
Zhejiang Provincial Natural Science Foundation of China (No.
Fig. 1 Cytokines derived from distinct subsets of CD4
+
T cells affect
anti-HBV ability of host immune response. Upon activation by antigen
presented cells, naive CD4
+
T cells commit to different T cell subsets
including Th1, Th2, Th17, and Tregs under certain stimulation
conditions; afterwards, these subsets of T cells are able to exert separate
functions by producing subset-specific cytokines, thereby playing crucial
roles during the process of anti-viral immunity inHBV infected liver. This
working model have described cytokines secreted by different subsets of
CD4
+
T cells can activate or inhibit the anti-viral ability by enhancing or
attenuating the function of CD8
+
T, CD4
+
T and B cells, which provides
perspicuous thought for further conception and design for HBV-targeted
immunotherapeutics
50 Clinic Rev Allerg Immunol (2016) 50:4154
LY14H030001), and the Major national S&T Projects for infectious
diseases (2012ZX10002002). We acknowledge Prof. M. Eric Gershwin
and the reviewers for the helpful and thoughtful comments for the
manuscript.
Conflict of Interest Xuefen Li, Xia Liu, Li Tian, and Yu Chen declare
that they have no conflict of interest.
References
1. Villa E, Fattovich G, Mauro A, Pasino M (2011) Natural history of
chronic HBV infection: special emphasis on the prognostic impli-
cations of the inactive carrier state versus chronic hepatitis. Dig
Liver Dis 43(Suppl 1):S8S14
2. Ganem D, Prince AM (2004) Hepatitis B virus infectionnatural
history and clinical consequences. N Engl J Med 350:11181129
3. Dienstag JL (2008) Hepatitis B virus infection. N Engl J Med 359:
14861500
4. Zhang Z, Zhang JY, Wang LF, Wang FS (2012)
Immunopathogenesis and prognostic immune markers of chronic
hepatitis B virus infection. J Gastroenterol Hepatol 27:223230
5. Chisari FV, Isogawa M, Wieland SF (2010) Pathogenesis of hepa-
titis B virus infection. Pathol Biol (Paris) 58:258266
6. Bauer T, Sprinzl M, Protzer U (2011) Immune control of hepatitis B
virus. Dig Dis 29:423433
7. Guidotti LG, Chisari FV (2001) Noncytolytic control of viral infec-
tions by the innate and adaptive immune response. Annu Rev
Immunol 19:6591
8. Fisicaro P, Valdatta C, Boni C et al (2009) Early kinetics of innate
and adaptive immune responses during hepatitis B virus infection.
Gut 58:974982
9. Uprichard SL, Wieland SF, Althage A, Chisari FV (2003)
Transcriptional and posttranscriptional control of hepatitis B virus
gene expression. Proc Natl Acad Sci U S A 100:13101315
10. Wieland SF, Guidotti LG, Chisari FV (2000) Intrahepatic induction
of alpha/beta interferon eliminates viral RNA-containing capsids in
hepatitis B virus transgenic mice. J Virol 74:41654173
11. He YL, Zhao YR, Zhang SL, Lin SM (2006) Host susceptibility to
persistent hepatitis B virus infection. World J Gastroenterol 12:
47884793
12. Zhang W, Yue B, Wang GQ, Lu SL (2002) Serum and ascites levels
of macrophage migration inhibitory factor, TNF-alpha and IL-6 in
patients with chronic virus hepatitis B and hepatitis cirrhosis.
Hepatobiliary Pancreat Dis Int 1:577580
13. Odeh M, Sabo E, Srugo I, Oliven A (2004) Serum levels of tumor
necrosis factor-alpha correlate with severity of hepatic encephalop-
athy due to chronic liver failure. Liver Int 24:110116
14. Xiao P, Chen QF, Yang YL, Guo ZH, Chen H (2006) Serum soluble
interleukin-2 receptor levels in patients with chronic hepatitis B
virus infection and its relation with anti-HBc. World J
Gastroenterol 12:482484
15. Liu M, Miao T, Zhu H et al (2012) IL-2-engineered nano-APC
effectively activates viral antigen-mediated T cell responses from
chronic hepatitis B virus-infected patients. J Immunol 188:1534
1543
16. Rossol S, Marinos G, Carucci P, Singer MV, Williams R, Naoumov
NV (1997) Interleukin-12 induction of Th1 cytokines is important
for viral clearance in chronic hepatitis B. J Clin Invest 99:3025
3033
17. le Song H, Binh VQ, Duy DN et al (2003) Serum cytokine profiles
associated with clinical presentation in Vietnamese infected with
hepatitis B virus. J Clin Virol 28:93103
18. Schurich A, Pallett LJ, Lubowiecki M et al (2013) The third signal
cytokine IL-12 rescues the anti-viral function of exhausted HBV-
specific CD8 T cells. PLoS Pathog 9:e1003208
19. Dadmanesh M, Ghorban K, Hassanshahi G, Momeni M, Arababadi
MK (2014) Current information concerning association of IL-12
and hepatitis B infection. Clin Lab 60:185191
20. He D, Yan G, Wang Y (2012) Serum levels of interleukin-12 in
various clinical states with hepatitis B virus infection. Cell Immunol
272:162165
21. Jegaskanda S, Ahn SH, Skinner N et al (2014) Down-regulation of
IL-18 mediated cell signalling and IFN-gamma expression by the
hepatitis B virus e antigen. J Virol 88:1041210420
22. Li W, Kubo S, Okuda A et al (2010) Effect of IL-18 on expansion of
gammadelta T cells stimulated by zoledronate and IL-2. J
Immunother 33:287296
23. Hegde S, Chen X, Keaton JM, Reddington F, Besra GS, Gumperz
JE (2007) NKT cells direct monocytes into a DC differentiation
pathway. J Leukoc Biol 81:12241235
24. Nelms K, Keegan AD, Zamorano J, Ryan JJ, Paul WE (1999) The
IL-4 receptor: signaling mechanisms and biologic functions. Annu
Rev Immunol 17:701738
25. Jiang Y, Ma Z, Xin G et al (2010) Th1 and Th2 immune response in
chronic hepatitis B patients during a long-term treatment with
adefovir dipivoxil. Mediat Inflamm 2010:143026
26. Yao Y, Li J, Lu Z et al (2011)Proteomic analysis of the interleukin-4
(IL-4) response in hepatitis B virus-positive human hepatocelluar
carcinoma cell line HepG2.2.15. Electrophoresis 32:20042012
27. Barathan M, Mariappan V, Shankar EM, Abdullah BJ, Goh KL,
Vadivelu J (2013) Hypericin-photodynamic therapy leads to
interleukin-6 secretion by HepG2 cells and their apoptosis via
recruitment of BH3 interacting-domain death agonist and caspases.
Cell Death Dis 4:e697
28. Missale G, Ferrari C, Fiaccadori F (1995) Cytokine mediators in
acute inflammation and chronic course of viral hepatitis. Ann Ital
Med Int 10:1418
29. Buke AC, Buke M, Altuglu IE et al (2004) Tumor necrosis factor
alpha and interleukin 6 productions in response to platelet-activating
factor in chronic hepatitis B virus infection. Med Princ Pract 13:
273276
30. Park Y, Han KH, Kim HS (2011) Serum cytokine levels in patients
with chronic hepatitis B according to lamivudine therapy. J Clin Lab
Anal 25:414421
31. Karimi-Googheri M, Daneshvar H, Nosratabadi R et al (2014)
Important roles played by TGF-beta in hepatitis B infection. J
Med Virol 86:102108
32. Hong MH, Chou YC, Wu YC et al (2012) Transforming growth
factor-beta1 suppresses hepatitis B virus replication by the reduction
of hepatocyte nuclear factor-4alpha expression. PLoS One 7:e30360
33. Park SO, Kumar M, Gupta S (2012) TGF-beta and iron differently
alter HBV replication in human hepatocytes through TGF-beta/BMP
signaling and cellular microRNA expression. PLoS One 7:e39276
34. Sun C, Fu B, Gao Y et al (2012) TGF-beta1 down-regulation of
NKG2D/DAP10 and 2B4/SAP expression on human NK cells
contributes to HBV persistence. PLoS Pathog 8:e1002594
35. Wu JF, Wu TC, Chen CH et al (2010) Serum levels of interleukin-10
and interleukin-12 predict early, spontaneous hepatitis B virus e
antigen seroconversion. Gastroenterology 138(165172):e161e163
36. Sabat R, Grutz G, Warszawska K et al (2010) Biology of interleu-
kin-10. Cytokine Growth Factor Rev 21:331344
37. Das A, Ellis G, Pallant C et al (2012) IL-10-producing regulatory B
cells in the pathogenesis of chronic hepatitis B virus infection. J
Immunol 189:39253935
38. Fenoglio D, Bernuzzi F, Battaglia F et al (2012) Th17 and regula-
tory T lymphocytes in primary biliary cirrhosis and systemic scle-
rosis as models of autoimmune fibrotic diseases. Autoimmun Rev
12:300304
Clinic Rev Allerg Immunol (2016) 50:4154 51
39. Wu W, Li J, Chen F, Zhu H, Peng G, Chen Z (2010) Circulating
Th17 cells frequency is associated with the disease progression in
HBV infected patients. J Gastroenterol Hepatol 25:750757
40. Kolls JK, Linden A (2004) Interleukin-17 family members and
inflammation. Immunity 21:467476
41. Zhao L, Tang Y, You Z et al (2011) Interleukin-17 contributes to the
pathogenesis of autoimmune hepatitis through inducing hepatic
interleukin-6 expression. PLoS One 6:e18909
42. Du WJ, Zhen JH, Zeng ZQ et al (2013) Expression of interleukin-17
associated with disease progression and liver fibrosis with hepatitis
B virus infection: IL-17 in HBV infection. Diagn Pathol 8:40
43. Huang Z, van Velkinburgh JC, Ni B, Wu Y (2012) Pivotal roles of
the interleukin-23/T helper 17 cell axis in hepatitis B. Liver Int 32:
894901
44. Langrish CL, McKenzie BS, Wilson NJ, de Waal MR, Kastelein
RA, Cua DJ (2004) IL-12 and IL-23: master regulators of innateand
adaptive immunity. Immunol Rev 202:96105
45. Wang Q, Zhou J, Zhang B et al (2013) Hepatitis B virus induces IL-
23 production in antigen presenting cells and causes liver damage
via the IL-23/IL-17 axis. PLoS Pathog 9:e1003410
46. Liang SC, Tan XY, Luxenberg DP et al (2006) Interleukin (IL)-22
and IL-17 are coexpressed by Th17 cells and cooperatively enhance
expression of antimicrobial peptides. J Exp Med 203:22712279
47. Eyerich S, Eyerich K, Pennino D et al (2009) Th22 cells represent a
distinct human T cell subset involved in epidermal immunity and
remodeling. J Clin Invest 119:35733585
48. Hughes T, Becknell B, McClory S et al (2009) Stage 3 immature
human natural killer cells found in secondary lymphoid tissue
constitutively and selectively express the TH 17 cytokine interleu-
kin-22. Blood 113:40084010
49. Radaeva S, Sun R, Pan HN, Hong F, Gao B (2004) Interleukin 22
(IL-22) plays a protective role in T cell-mediated murine hepatitis:
IL-22 is a survival factor for hepatocytes via STAT3 activation.
Hepatology 39:13321342
50. Park O, Wang H, Weng H et al (2011) In vivo consequences of liver-
specific interleukin-22 expression in mice: implications for human
liver disease progression. Hepatology 54:252261
51. Zhang Y, Cobleigh MA, Lian JQ et al (2011) A proinflammatory
role for interleukin-22 in the immune response to hepatitis B virus.
Gastroenterology 141:18971906
52. Monteleone G, Pallone F, Macdonald TT (2009) Interleukin-21 (IL-
21)-mediated pathways in T cell-mediated disease. Cytokine
Growth Factor Rev 20:185191
53. Spolski R, Leonard WJ (2008) The Yin and Yang of interleukin-21 in
allergy, autoimmunity and cancer. Curr Opin Immunol 20:295301
54. Spolski R, Leonard WJ (2010) IL-21 and T follicular helper cells.
Int Immunol 22:712
55. Publicover J, Goodsell A, Nishimura S et al (2011) IL-21 is pivotal
in determining age-dependent effectiveness of immune responses in
a mouse model of human hepatitis B. J Clin Invest 121:11541162
56. Franks I (2011) Viral hepatitis: interleukin 21 has a key role in age-
dependent response to HBV. Nat Rev Gastroenterol Hepatol 8:243
57. Tuncbilek S (2014) Relationship between cytokine gene polymor-
phisms and chronic hepatitis B virus infection. World J
Gastroenterol 20:62266235
58. Sodsai P, Surakiatchanukul T, Kupatawintu P, Tangkitvanich P,
Hirankarn N (2013) Association of cytokine and cytokine receptor
gene polymorphisms with the risk of chronic hepatitis B. Asian Pac
J Allergy Immunol 31:277285
59. Lian JQ, Yang XF, Zhao RR et al (2014) Expression profiles of
circulating cytokines, chemokines and immune cells in patients with
hepatitis B virus infection. Hepat Mon 14:e18892
60. Abbas Z, Siddiqui AR (2011) Management of hepatitis B in devel-
oping countries. World J Hepatol 3:292299
61. Liver EAFTSOT (2012) EASL clinical practice guidelines: manage-
ment of chronic hepatitis B virus infection. J Hepatol 57:167185
62. Arababadi MK, Pourfathollah AA, Jafarzadeh A, Hassanshahi G
(2010) Serum levels of IL-10 and IL-17A in occult HBV-infected
South-East Iranian patients. Hepat Mon 10:3135
63. Zhang JY, Zhang Z, Lin F et al (2010) Interleukin-17-producing
CD4(+) T cells increase with severity of liver damage in patients
with chronic hepatitis B. Hepatology 51:8191
64. He D, Li M, Guo S et al (2013) Expression pattern of serum
cytokines in hepatitis B virus infected patients with persistently
normal alanine aminotransferase levels. J Clin Immunol 33:1240
1249
65. Bertolino P, McCaughan GW, Bowen DG (2002) Role of primary
intrahepatic T-cell activation in the liver tolerance effect.Immunol
Cell Biol 80:8492
66. Dumortier J, Schonig K, Oberwinkler H et al (2005) Liver-specific
expression of interferon gamma following adenoviral gene transfer
controls hepatitis B virus replication in mice. Gene Ther 12:668677
67. Isogawa M, Robek MD, Furuichi Y, Chisari FV (2005) Toll-like
receptor signaling inhibits hepatitis B virus replication in vivo. J
Virol 79:72697272
68. Chokshi S, Cooksley H, Riva A et al (2014) Identification of serum
cytokine profiles associated with HBeAg seroconversion following
antiviral treatment interruption. Viral Immunol 27:235244
69. Zhang G, Li Z, Han Q et al (2011) Altered TNF-alpha and IFN-gamma
levels associated with PD1 but not TNFA polymorphisms in patients
with chronic HBV infection. Infect Genet Evol 11:16241630
70. Wang B, Wang J, Zheng Y et al (2010) A study of TNF-alpha-238
and 308 polymorphisms with different outcomes of persistent
hepatitis B virus infection in China. Pathology 42:674680
71. Basturk B, Karasu Z, Kilic M, Ulukaya S, Boyacioglu S, Oral B
(2008) Association of TNF-alpha 308 polymorphism with the out-
come of hepatitis B virus infection in Turkey. Infect Genet Evol 8:20
25
72. Fletcher GJ, Samuel P, Christdas J et al (2011) Association of HLA
and TNF polymorphisms with the outcome of HBVinfection in the
South Indian population. Genes Immun 12:552558
73. Toe JG, Pellegrini M, Mak TW (2013) Promoting immunity during
chronic infectionthe therapeutic potential of common gamma-
chain cytokines. Mol Immunol 56:3847
74. Zeuzem S, Carreno V (2001) Interleukin-12 in the treatment of
chronic hepatitis B and C. Antivir Res 52:181188
75. Tung NH, Quang TH, Son JH et al (2011) Inhibitory effect of
ginsenosides from steamed ginseng-leaves and flowers on the
LPS-stimulated IL-12 production in bone marrow-derived dendritic
cells. Arch Pharm Res 34:681685
76. Gao QJ, Liu DW, Zhang SY et al (2009) Polymorphisms of some
cytokines and chronic hepatitis B and C virus infection. World J
Gastroenterol 15:56105619
77.DongY,XiH,YuY,WangQ,JiangK,LiL(2002)Effectsof
oxymatrine on the serum levels of T helper cell 1 and 2 cytokines
and the expression of the S gene in hepatitis B virus S gene
transgenic mice: a study on the anti-hepatitis B virus mechanism
of oxymatrine. J Gastroenterol Hepatol 17:12991306
78. Gur A, Dikici B, Nas K, Bosnak M, Haspolat K, Sarac AJ (2005)
Bone mineral density and cytokine levels during interferon therapy
in children with chronic hepatitis B: does interferon therapy prevent
from osteoporosis? BMC Gastroenterol 5:30
79. Kao JT, Lai HC, Tsai SM et al (2012) Rather than interleukin-27,
interleukin-6 expresses positive correlation with liver severity in
naive hepatitis B infection patients. Liver Int 32:928936
80. Pillai MR, Collison LW, Wang X et al (2011) The plasticity of
regulatory T cell function. J Immunol 187:49874997
81. Aalaei-Andabili SH, Alavian SM (2012) Regulatory T cells are the
most important determinant factor of hepatitis B infection progno-
sis: a systematic review and meta-analysis. Vaccine 30:55955602
82. Xu D, Fu J, Jin L et al (2006) Circulating and liver resident CD4
+
CD25
+
regulatory T cells actively influence the antiviral immune
52 Clinic Rev Allerg Immunol (2016) 50:4154
response and disease progression in patients with hepatitis B. J
Immunol 177:739747
83. Liu F, Tong F, He Y, Liu H (2011) Detectable expression of IL-35 in
CD4
+
T cells from peripheral blood of chronic hepatitis B patients.
Clin Immunol 139:15
84. Zhang KQ, Polga D, Salzman SA, Burmester JK (2002) Amino
acids 67 and 68 of transforming growth factor-beta regulate binding
to a glycosyl phosphatidyl inositol-linked membrane protein on
vascular endothelial cells. Cytokines Cell Mol Ther 7:2530
85. Besnard AG, Sabat R, Dumoutier L et al (2011) Dual Role of IL-22
in allergic airway inflammation and its cross-talk with IL-17A. Am
J Respir Crit Care Med 183:11531163
86. Chen Y, Xie X, Gu L, Huang XH, Peng XM (2013) Furin mRNA
expression in peripheral blood correlates with chronic hepatitis B
virus infection. Hepatol Res 43:208216
87. Fiorentino DF, Bond MW, Mosmann TR (1989) Two types of
mouse T helper cell. IV. Th2 clones secrete a factor that inhibits
cytokine production by Th1 clones. J Exp Med 170:20812095
88. Ejrnaes M, Filippi CM, Martinic MM et al (2006) Resolution of a
chronic viral infection after interleukin-10 receptor blockade. J Exp
Med 203:24612472
89. Wang B, Zhao XP, Fan YC, Zhang JJ, Zhao J, Wang K (2013) IL-
17A but not IL-22 suppresses the replication of hepatitis B virus
mediated by over-expression of MxA and OAS mRNA in the
HepG2.2.15 cell line. Antiviral Res 97:285292
90. Oppmann B, Lesley R, Blom B et al (2000) Novel p19 protein
engages IL-12p40 to form a cytokine, IL-23, with biological activ-
ities similar as well as distinct from IL-12. Immunity 13:715725
91. Xie MH, Aggarwal S, Ho WH et al (2000) Interleukin (IL)-22, a
novel human cytokine that signals through the interferon receptor-
related proteins CRF2-4 and IL-22R. J Biol Chem 275:3133531339
92. Kotenko SV, Izotova LS, Mirochnitchenko OV et al (2001)
Identification of the functional interleukin-22 (IL-22) receptor com-
plex: the IL-10R2 chain (IL-10Rbeta ) is a common chain of both
the IL-10 and IL-22 (IL-10-related T cell-derived inducible factor,
IL-TIF) receptor complexes. J Biol Chem 276:27252732
93. Brand S, Dambacher J, Beigel F et al (2007) IL-22-mediated liver
cell regeneration is abrogated by SOCS-1/3 overexpression in vitro.
Am J Physiol Gastrointest Liver Physiol 292:G1019G1028
94. Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy AJ,
Karow M, Flavell RA (2007) Interleukin-22 but not interleukin-17
provides protection to hepatocytes during acute liver inflammation.
Immunity 27:647659
95. Kakimi K, Lane TE, Wieland S et al (2001) Blocking chemokine
responsive to gamma-2/interferon (IFN)-gamma inducible protein
and monokine induced by IFN-gamma activity in vivo reduces the
pathogenetic but not the antiviral potential of hepatitis B virus-
specific cytotoxic T lymphocytes. J Exp Med 194:17551766
96. Xiang X, Gui H, King NJ et al (2012) IL-22 and non-ELR-CXC
chemokine expression in chronic hepatitis B virus-infected liver.
ImmunolCellBiol90:611619
97. Zeng R, Spolski R, Casas E, Zhu W, Levy DE, Leonard WJ (2007)
The molecular basis of IL-21-mediated proliferation. Blood 109:
41354142
98. Spolski R, Leonard WJ (2008) Interleukin-21: basic biology and im-
plications for cancer and autoimmunity. Annu Rev Immunol 26:5779
99. Ettinger R, Kuchen S, Lipsky PE (2008) The role of IL-21 in
regulating B-cell function in health and disease. Immunol Rev 223:
6086
100. Iannello A, Tremblay C, Routy JP, Boulassel MR, Toma E, Ahmad
A (2008) Decreased levels of circulating IL-21 in HIV-infected
AIDS patients: correlation with CD4
+
T-cell counts. Viral
Immunol 21:385388
101. Hu X, Ma S, Huang X et al (2011) Interleukin-21 is upregulated in
hepatitis B-related acute-on-chronic liver failure and associated with
severity of liver disease. J Viral Hepat 18:458467
102. Kwon H, Lok AS (2011) Hepatitis B therapy. Nat Rev Gastroenterol
Hepatol 8:275284
103. Tseng TC, Kao JH, Chen DS (2014) Peginterferon alpha in the
treatment of chronic hepatitis B. Expert Opin Biol Ther 14:9951006
104. Liu YZ, Hou FQ, Ding P, Ren YY, Li SH, Wang GQ (2012)
Pegylated interferon alpha enhances recovery of memory T cells
in e antigen positive chronic hepatitis B patients. Virol J 9:274
105. Belloni L, Allweiss L, Guerrieri F et al (2012) IFN-alpha inhibits
HBV transcription and replication in cell culture and in humanized
mice by targeting the epigenetic regulation of the nuclear cccDNA
minichromosome. J Clin Invest 122:529537
106. Xu C, Guo H, Pan XB et al (2010) Interferons accelerate decay of
replication-competent nucleocapsids of hepatitis B virus. J Virol 84:
93329340
107. Brunetto MR, Oliveri F, Colombatto P, Coco B, Ciccorossi P,
Bonino F (2003) Treatment of HBeAg-negative chronic hepatitis
B with interferon or pegylated interferon. J Hepatol 39(Suppl 1):
S164S167
108. Rigopoulou EI, Suri D, Chokshi S et al (2005) Lamivudine plus
interleukin-12 combination therapy in chronic hepatitis B: antiviral
and immunological activity. Hepatology 42:10281036
109. Szkaradkiewicz A, Jopek A, Wysocki J (2005) Effects of IL-12 and IL-
18 on HBcAg-specific cytokine production by CD4 T lymphocytes of
children with chronic hepatitis B infection. Antiviral Res 66:2327
110. Grimm D, Heeg M, Thimme R (2013) Hepatitis B virus: from
immunobiology to immunotherapy. Clin Sci (Lond) 124:7785
111. Ghany MG, Doo EC (2009) Antiviral resistance and hepatitis B
therapy. Hepatology 49:S174S184
112. Nebbia G, Peppa D, Maini MK (2012) Hepatitis B infection: current
concepts and future challenges. QJM 105:109113
113. Ahlenstiel G, Edlich B, Hogdal LJ et al (2011) Early changes in
natural killer cell function indicate virologic response to interferon
therapy for hepatitis C. Gastroenterology 141:12311239, 1239
e1231-1232
114. Craxi A, Di Bona D, Camma C (2003) Interferon-alpha for HBeAg-
positive chronic hepatitis B. J Hepatol 39(Suppl 1):S99S105
115. Shi B, Ren G, Hu Y, Wang S, Zhang Z, Yuan Z (2012) HBsAg
inhibits IFN-alpha production in plasmacytoid dendritic cells
through TNF-alpha and IL-10 induction in monocytes. PLoS One
7:e44900
116. Walsh R, Locarnini S (2012) Hepatitis B precore protein: pathogen-
ic potential and therapeutic promise. Yonsei Med J 53:875885
117. Reijnders JG, Perquin MJ, Zhang N, Hansen BE, Janssen HL
(2010) Nucleos(t)ide analogues only induce temporary hepatitis B
e antigen seroconversion in most patients with chronic hepatitis B.
Gastroenterology 139:491498
118. Buster EH, Hansen BE, Lau GK et al (2009) Factors that predict
response of patients with hepatitis B e antigen-positive chronic
hepatitis B to peginterferon-alfa. Gastroenterology 137:20022009
119. Janssen HL, van Zonneveld M, Senturk H et al (2005) Pegylated
interferon alfa-2b alone or in combination with lamivudine for
HBeAg-positive chronic hepatitis B: a randomised trial. Lancet
365:123129
120. Lau GK, Piratvisuth T, Luo KX et al (2005) Peginterferon Alfa-2a,
lamivudine, and the combination for HBeAg-positive chronic hep-
atitis B. N Engl J Med 352:26822695
121. Piccolo P, Lenci I, Demelia L et al (2009) A randomized controlled
trial of pegylated interferon-alpha2a plus adefovir dipivoxil for
hepatitis B e antigen-negative chronic hepatitis B. Antivir Ther 14:
11651174
122. Takkenberg B, Terpstra V, Zaaijer H et al (2011) Intrahepatic re-
sponse markers in chronic hepatitis B patients treated with
peginterferon alpha-2a and adefovir. J Gastroenterol Hepatol 26:
15271535
123. Scott DL, Wolfe F, Huizinga TW (2010) Rheumatoid arthritis.
Lancet 376:10941108
Clinic Rev Allerg Immunol (2016) 50:4154 53
124. Rutgeerts P, Vermeire S, Van Assche G (2009) Biological therapies
for inflammatory bowel diseases. Gastroenterology 136:11821197
125. Ostuni P, Botsios C, Punzi L, Sfriso P, Todesco S (2003) Hepatitis B
reactivation in a chronic hepatitis B surface antigen carrier with
rheumatoid arthritis treated with infliximab and low dose metho-
trexate. Ann Rheum Dis 62:686687
126. Chevaux JB, Nani A, Oussalah A et al (2010) Prevalence of hepatitis
B and C and risk factors for nonvaccination in inflammatory bowel
disease patients in Northeast France. Inflamm Bowel Dis 16:916924
127. Robinson H, Walker-Bone K (2009) Anti-TNF-alpha therapy for
rheumatoid arthritis among patients with chronic hepatitis B infec-
tion. Rheumatology (Oxford) 48:448450
128. Ryu HH, Lee EY, Shin K et al (2012) Hepatitis B virus reactivation
in rheumatoid arthritis and ankylosing spondylitis patients treated
with anti-TNFalpha agents: a retrospective analysis of 49 cases. Clin
Rheumatol 31:931936
129. Lee YH, Bae SC, Song GG (2013) Hepatitis B virus reactivation in
HBsAg-positive patients with rheumatic diseases undergoing anti-tumor
necrosis factor therapy or DMARDs. Int J Rheum Dis 16:527531
130. Vassilopoulos D (2011) Should we routinely treat patients with
autoimmune/rheumatic diseases and chronic hepatitis B virus infec-
tion starting biologic therapies with antiviral agents? Yes. Eur J
Intern Med 22:572575
131. Nosotti L, Francesconi F, Izzi S, Berardesca E, Morrone A, Bonifati
C (2010) Safety of antitumour necrosis factor-alpha therapy in
psoriatic patients with hepatitis B virus infection. Br J Dermatol
162:14081410
132. Arababadi MK, Pourfathollah AA, Jafarzadeh A et al (2011) Non-
association of IL-12 +1188 and IFN-gamma +874 polymorphisms
with cytokines serum level in occult HBVinfected patients. Saudi J
Gastroenterol 17:3035
133. Pramoolsinsup C (2002) Management of viral hepatitis B. J
Gastroenterol Hepatol 17(Suppl):S125S145
134. Carreno V, Zeuzem S, Hopf U et al (2000) A phase I/II study of
recombinant human interleukin-12 in patients with chronic hepatitis
B. J Hepatol 32:317324
135. Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB (2004)
Interleukin-10 and related cytokines and receptors. Annu Rev
Immunol 22:929979
136. Goddard S, Youster J, Morgan E, Adams DH (2004) Interleukin-10
secretion differentiates dendritic cells from human liver and skin.
Am J Pathol 164:511519
137. Tjwa ET, van Oord GW, Hegmans JP, Janssen HL, Woltman AM
(2011) Viral load reduction improves activation and function of natural
killer cells in patients with chronic hepatitis B. J Hepatol 54:209218
138. Chou YC, Chen ML, Hu CP et al (2007) Transforming growth
factor-beta1 suppresses hepatitis B virus replication primarily
through transcriptional inhibition of pregenomic RNA.
Hepatology 46:672681
139. Tarr PE, Lin R, Mueller EA, Kovarik JM, Guillaume M, Jones TC
(1996) Evaluation of tolerability and antibody response after
recombinant human granulocyte-macrophage colony-stimulating
factor (rhGM-CSF) and a single dose of recombinant hepatitis B
vaccine. Vaccine 14:11991204
140. Martin J, Bosch O, Moraleda G, Bartolome J, Quiroga JA, Carreno
V (1993) Pilot study of recombinant human granulocyte-
macrophage colony-stimulating factor in the treatment of chronic
hepatitis B. Hepatology 18:775780
141. Wang J, Zhu Q, Zhang T, Yu H (2002) A pilot study on the
combined therapy of granulocyte-macrophage colony-stimulating
factor and hepatitis B vaccine on chronic hepatitis B virus carrier
children. Chin Med J (Engl) 115:18241828
142. Qing Y, Chen M, Zhao J et al (2010) Constructionof an HBV DNA
vaccine by fusion of the GM-CSF gene to the HBV-S gene and
examination of its immune effects in normal and HBV-transgenic
mice. Vaccine 28:43014307
143. Overton ET, Sungkanuparph S, Klebert M et al (2011) GM-CSF
fails to improve immune responses to booster hepatitis B vaccina-
tion in HIV-infected individuals. Open Virol J 5:109113
144. Yuan Q, Li PD, Li BH et al (2009) Differential IL-4/Stat6 activities
correlate with differential expression of regulatory genes SOCS-1,
SHP-1, and PP2A in colon cancer cells. J Cancer Res Clin Oncol
135:131140
145. Corry DB, Kheradmand F (2002) Biology and therapeutic potential
of the interleukin-4/interleukin-13 signaling pathway in asthma. Am
J Respir Med 1:185193
146. Fan Z, Spencer JA, Lu Y et al (2010) In vivo tracking of color-
codedeffector, natural and induced regulatory T cells in the allo-
graft response. Nat Med 16:718722
147. Meyers JA, Liu AY, McTiernan A et al (2008) Serum leptin
concentrations and markers of immune function in over-
weight or obese postmenopausal women. J Endocrinol 199:
5160
148. Gorski KS, Waller EL, Bjornton-Severson J et al (2006) Distinct
indirect pathways govern human NK-cell activation by TLR-7 and
TLR-8 agonists. Int Immunol 18:11151126
149. Wilson MT, Johansson C, Olivares-Villagomez D et al (2003) The
response of natural killer T cells to glycolipid antigens is character-
ized by surface receptor down-modulation and expansion. ProcNatl
Acad Sci U S A 100:1091310918
150. Comella K, Nakamura M, Melnik K et al (2001) Effects of antibody
concentration on the separation of human natural killer cells in a
commercial immunomagnetic separation system. Cytometry 45:
285293
151. Matsuda JL, Naidenko OV, Gapin L et al (2000) Tracking the
response of natural killer T cells to a glycolipid antigen using
CD1d tetramers. J Exp Med 192:741754
152. Wynn TA (2004) Fibrotic disease and the T(H)1/T(H)2 paradigm.
Nat Rev Immunol 4:583594
153. Meier R, Golovko D, Tavri S et al (2011) Depicting adoptive
immunotherapy for prostate cancer in an animal model with mag-
netic resonance imaging. Magn Reson Med 65:756763
54 Clinic Rev Allerg Immunol (2016) 50:4154
... Therefore, serological detection of HBV-related liver disease is critical for monitoring progression and determining prognosis. Previous studies have investigated the role of serum cytokine detection in HBV-associated liver disease [9,10]. Most related studies have focused on the effect of only a single cytokine, but the regulatory network of cytokines is highly complex [2,11]. ...
... HBV poses a severe threat to public health and is a high risk factor for chronic hepatitis, occult hepatitis, cirrhosis, and HCC [1,14]. As HBV is a noncytopathic virus, the associated inflammation and direct liver damage caused by this virus are mediated by the immune response [9]. Understanding the expression levels of serum cytokines is crucial for understanding the development and metastasis of tumors and is helpful for determining the prognosis and treatment of tumors [7,15,16]. ...
... Therefore, Luminex liquid chip analysis was used in this study because this technology has high sensitivity and can simultaneously detect ten serum cytokines associated with HBV infection [11]. In addition, compared with solid-phase chip technology, the Luminex liquid chip has the advantages of a simple procedure, good repeatability, and high throughput [9,17]. ...
Article
Full-text available
Background Cytokines/chemokines play essential roles in the occurrence and progression of hepatitis B virus (HBV) infection. This study aimed to observe the expression patterns of 10 related cytokines/chemokines in the serum of healthy individuals, self-limited patients and HBV-infected patients at different stages of disease (chronic hepatitis B (CHB), liver cirrhosis (LC), hepatocellular dysplastic nodules (DNs) and hepatocellular carcinoma (HCC)) and to analyze the relationships of these cytokines/chemokines with disease progression. Methods The levels of six cytokines (FGF-2, IFN-α2, IL-4, IL-6, IL-10 and VEGF-A) and four chemokines (GRO-α, IL-8, IP-10 and MCP-1) were quantified using Luminex multiplex technology. Results There were no significant differences in the expression of the 10 cytokines/chemokines between healthy individuals and self-limited patients. The levels of IL-4, IL-6, and IL-8 increased significantly in the CHB and LC groups. IL-10 was highly expressed in the HCC group. The level of IP-10 was significantly greater in all liver disease groups (CHB, LC, DN and HCC) than in the HI and SL-HBV groups, while the level of GRO was significantly lower in all liver disease groups than in the HI and SL-HBV groups. The levels of the 10 cytokines/chemokines were not significantly different between the preoperative group and the two-day postoperative group. Significant increases in the levels of IL-4, VEGF-A and IL-8 and significant decreases in those of IL-10 and GRO-α were observed 3 months after surgery. Correlation analysis revealed that most of the cytokines/chemokines with significant correlation differences were positively correlated before and after HCC surgery. Conclusion Our results highlight the fluctuating status of specific cytokines in HBV infection-related disease progression. It is speculated that these cytokines may be used as serum markers to monitor dynamic changes during the progression of HBV-related liver disease and to predict patient prognosis.
... Thus, both arms of the immune system are involved in disease manifestation or long-lasting protection [14]. Therefore, appropriate Th1 immunity and cytokine network are vital in developing protective immune responses against HBV [15]. However, inappropriate host inflammatory responses in the liver are implicated in the pathogenesis of fulminant, acute, chronic or HCC caused by HBV infection. ...
... Some studies suggest that Th1 should be the main dual-function host cell for HBV infection [15,32]. On the one hand, its intense and uncontrolled activity causes type IV hypersensitivity, which plays a significant role in fulminant and acute hepatitis and induction of cirrhosis. ...
Article
The hepatitis B virus (HBV) infection has a wide range, from fulminant hepatitis to inactive chronic hepatitis B (ICB) infection. The present study evaluated critical factors in the outcomes of HBV infection in a highly endemic region of Iran (approximately 12% HBV positive). The expression of seven genes involved in host immunity (Foxp3, T-bet, ROR-γt, AKT, CREB, IL-28/or IFN-λ2, and IL-28R) and HBx for viral activities were evaluated using real-time PCR, TaqMan method. A total of 58 subjects were randomly chosen, including 28 ICB and 30 healthy controls (HCs) from the Esfandiar district, South Khorasan province, Iran. The expression index of Foxp3 and ROR-γt was moderately up-regulated in ICBs but did not statistically significant. T-bet expression in ICB patients was significantly higher than in HCs (p = 0.004). Furthermore, evaluating two signalling pathways in Th activation and cell survival showed that the CREB pathway was significantly up-regulated in ICB patients compared to HCs (p = 0.006), but the AKT did not differ. In innate immune responses, the IL-28/or IFN-λ2 expression in ICB patients was significantly higher than in the HCs (p = 0.02). Surprisingly, only one ICB patient disclosed HBx expression, which shows deficient virus activity in these patients. The ICB condition seems to result from host immune pressure on HBV activities, up-regulation of T-bet and IFN-λ. The high expression of CREB may prevent Kupffer's pro-inflammatory reactions in the liver. Whereas the absence of HBx expression in ICB patients and, consequently, the inactivity of HBV may also confirm such immune pressure.
... (2) Dysregulation of cytokines: ICIs can cause a dysregulation of cytokines, which are proteins that regulate the immune response. This dysregulation can lead to an overactive immune response against liver cells [87,88]. (3) Direct toxicity: Some ICIs may have direct toxic effects on liver cells, leading to hepatitis [89][90][91]. ...
Article
Full-text available
Immune checkpoint inhibitors (ICIs) have generated considerable excitement as a novel class of immunotherapeutic agents due to their remarkable efficacy in treating various types of cancer. However, the widespread use of ICIs has brought about a number of safety concerns, especially the development of immune-related adverse events (irAEs). These serious complications could result in treatment discontinuation and even life-threatening consequences, making it critical to identify high-risk groups and predictive markers of irAEs before initiating therapy. To this end, the current article examines several potential predictive markers of irAEs in important organs affected by ICIs. While retrospective studies have yielded some promising results, limitations such as small sample sizes, variable patient populations, and specific cancer types and ICIs studied make it difficult to generalize the findings. Therefore, prospective cohort studies and real-world investigations are needed to validate the potential of different biomarkers in predicting irAEs risk. Overall, identifying predictive markers of irAEs is a crucial step towards improving patient safety and enhancing the management of irAEs. With ongoing research efforts, it is hoped that more accurate and reliable biomarkers will be identified and incorporated into clinical practice to guide treatment decisions and prevent the development of irAEs in susceptible patients.
... Apart from being a main mediator of inflammatory processes, IL-6 is also involved in homeostatic liver mechanisms. In fact, it promotes liver regeneration and protects liver cells from injuries caused by immune responses, alcohol, and viral infections [49,50]. Moreover, IL-6 seems to play a crucial role in protecting against HBV infection. ...
Article
Full-text available
Objective The objective of this study was to assess the possibility of HBV reactivation (HBVr) in patients with RA under anti-IL-6 treatment. Methods Using PubMed, Scopus and EMBASE, we performed a systematic literature search for articles related to HBVr in RA patients under anti-IL-6 treatment. The search was performed with no date limits and was last updated 28 January 2023. The results from all the databases were combined and duplicates were excluded, as were non-English articles, case reports, position articles, comments, and paediatric studies. Results Our initial search led to 427 articles; 28 were duplicates, 46 non-English, 169 reviews, 31 books/letters, 25 case reports, and 88 irrelevant to the meta-analysis aim; 21 were excluded due to inadequate information, leaving 19 articles, with a sum of 372 RA patients with chronic HBV (CHB) or resolved HBV infection, for further analysis. The overall risk for HBVr in RA patients with CHB was 6.7%, increasing to 37% when only RA patients with CHB and no antiviral prophylaxis were included. On the contrary, HBVr was close to 0% in RA patients with resolved HBV infection, irrespective of antiviral prophylaxis. All RA patients experiencing HBVr in these studies were successfully managed with antiviral treatment and/or drug withdrawal. Conclusion Overall, anti-IL-6 treatment comes with a significant risk of HBVr in RA patients with CHB; risk is diminished when antiviral prophylaxis is used. In contrast, in RA patients with resolved HBV infection, the risk of HBVr seems to be extremely low. Large, well-designed studies (either controlled trials or multicentre/international observational studies) are warranted to further validate these results.
... Nhiễm vi rút viêm gan B (HBV) là một bệnh lý truyền nhiễm phổ biến trên thế giới, với khoảng 1/3 dân số nhiễm bệnh. Đáp ứng qua trung gian miễn dịch, điều hòa các con đường tín hiệu cytokine đã được chứng minh có vai trò quan trọng trong cơ chế bệnh sinh và kết cục lâm sàng của nhiễm vi rút viêm gan B [1]. ...
Article
Mục tiêu: Xây dựng quy trình phát hiện điểm đa hình đơn nucleotide (SNP) rs2062345 gen SOCS6 trên bệnh nhân mang virus viêm gan B bằng phương pháp Tetra-primer ARMS PCR (T-ARMS PCR). Đối tượng và phương pháp: Mẫu máu tĩnh mạch bệnh nhân mang virus viêm gan B (viêm gan, xơ gan, ung thư gan); sử dụng phương pháp T-ARMS PCR, có đối chiếu một số mẫu đại diện với phương pháp giải trình tự gen Sanger. Kết quả: Đã tối ưu được quy trình phản ứng T-ARMS PCR nhằm phát hiện SNP rs2062345 gen SOCS6; các kết quả điện di PCR đều rõ nét, không xuất hiện băng phụ phù hợp với kiểu gen của SNP rs2062345 gen SOCS6. Kết quả thí nghiệm T-ARMS PCR hoàn toàn tương đồng với phương pháp giải trình tự gen Sanger. Kết luận: Đã xây dựng thành công quy trình xác định SNP rs2062345 trên gen SOCS6 bằng phương pháp T-ARMS PCR.
... 9,10 The Role of Cytokine Against HBV Infection By binding to specific receptors expressed on the target cells, cytokines limit viral replication directly or indirectly. 11 Interleukin (IL)-6 and IL-1β regulate sodium-taurocholate cotransporting polypeptide (NTCP) expression and prevent HBV from entering cells. According to a recent study, cells pretreated with IL-6 reduced HBV entrance by up to 90%, significantly reducing the release of cccDNA and HBsAg. ...
Article
Full-text available
The antigen recognition and immune response to acute and chronic hepatitis B virus (HBV) infections are the result of both the innate and adaptive immune response. The innate immune response comprises Dendritic Cells (DCs), which served as professional antigen-presenting cells and a bridge between innate and adaptive immunity, Kupffer cells and inflammatory monocytes for the continuous inflammation of hepatocyte, neutrophils for hepatic tissue damage due to acute inflammation, type I interferons (IFN), which induce an antiviral state on infected cells, directs natural killer (NK) cells to kill virally infected cells, reduces the population of infected cells, and promotes the effective maturation and site recruitment of adaptive immunity through the production of pro-inflammatory cytokines and chemokines. Through stimulating B cells, T-helper, and cytotoxic T cells, the adaptive immune system also protects against hepatitis B infection. During HBV infection, a network of cell types that can either play protective or harmful functions creates the anti-viral adaptive immune response. These many elements, such as ‎Cluster of differentiation four (CD4) T cells (traditionally known as helper T cells), are potent cytokine producers and necessary for the effective maturation of effector cytotoxic cluster of differentiation eight (CD8) T cells and B cell antibody production. By cytolytic and non-cytolytic processes, CD8 T cells are able to eliminate HBV-infected hepatocytes and directly detect virus-infected cells, and circulating CD4+ CD25+ regulatory T cells for the modulation of immune system. In order to avoid reinfection, B cells can produce antibodies that destroy free viral particles. Moreover, by presenting HBV antigens to helper T cells, B cells may also influence how well these cells operate.
Article
While dysfunctional exhausted CD8+ T cells hamper viral control when children acquire hepatitis B virus (HBV) infection, it's crucial to recognize that CD8+ T cells have diverse phenotypes and functions. This study explored a subset of CD8+ T cells expressing C‐C chemokine receptor type 5 (CCR5) in children with HBV infection. Thirty‐six patients in the immune tolerant group, 33 patients in the immune active group, 55 patients in the combined response group, and 22 healthy control children were enrolled. The frequency, functional molecules, and effector functions of the CCR5+CD8+ T cell population in different groups were evaluated. The frequency of CCR5+CD8+ T cells correlated positively with the frequency of CCR5+CD4+ T cells and patient age, and it correlated negatively with alanine aminotransferase, aspartate transaminase, HBV DNA, hepatitis B surface antigen, and lactic dehydrogenase levels. CCR5+CD8+ T cells had higher levels of inhibitory and activated receptors and produced higher levels of IFN‐γ, IL‐2, and TNF‐α than CCR5‐CD8+ T cells. CCR5+CD8+ T cells were partially exhausted but possessed a stronger antiviral activity than CCR5‐CD8+ T cells. The identification of this subset increases our understanding of CD8+ T cell functions and serves as a potential immunotherapeutic target for children with HBV infection.
Article
Full-text available
Hepatitis B virus (HBV) infection is a worldwide medical issue with significant morbidity and mortality, as it is the main cause of chronic liver disease and hepatocellular carcinoma (HCC). Both innate and adaptive immune responses play a key role in HBV replication and suppression. Recently, the pathophysiological function of interleukins (IL) in the natural course of HBV has gained much attention as a result of the broad use of anti-interleukin agents for a variety of autoimmune diseases and the accompanying risk of HBV reactivation. We present a narrative review regarding the role of IL in HBV infection. Collectively, the pro-inflammatory ILs, namely IL-1, IL-5, IL-6, IL-12 and IL-21, seem to play a critical role in the suppression of HBV replication. In contrast, the anti-inflammatory cytokines IL-10, IL-23 and IL-35 probably act as HBV replication enhancers, while IL-17 has been correlated with HBV-related liver injury. Interestingly enough, IL-2, IL-4 and IL-12 have been tried as therapeutic options against HBV infection with contradictory results. Lastly, the role of IL-22 remains largely ill defined, although preliminary data suggest that it may play a significant role in HBV replication, proliferation and subsequent liver damage.
Article
Aims: Interleukin-34 (IL-34) and macrophage colony-stimulating factor (CSF-1) have similar functions, such as promoting the formation of liver fibrosis. This study aimed to evaluate and compare the diagnostic value of serum IL-34 and CSF-1 for significant liver fibrosis in patients with chronic hepatitis B (CHB). Methods: A total of 369 CHB patients, consisting of 208 HBeAg-negative patients and 161 HBeAg-positive patients, were enrolled in this study. Additionally, 72 healthy individuals served as healthy controls (HCs). Serum levels of IL-34 and CSF-1 were measured using the enzyme-linked immunosorbent assay method. Liver fibrosis grades were assessed using the modified Scheuer scoring system. Results: Serum IL-34 and CSF-1 levels exhibited significant elevation in both HBeAg-negative and HBeAg-positive patients in comparison to HCs (p < 0.001). IL-34 emerged as an independent factor linked to significant liver fibrosis, whereas CSF-1 did not exhibit such an association. Receiver operating characteristic (ROC) analysis indicated higher areas under the curves (AUCs) for IL-34 (0.814, p < 0.001 and 0.673, p < 0.001) when diagnosing significant liver fibrosis in HBeAg-negative and HBeAg-positive patients, respectively, as opposed to CSF-1 (0.602, p < 0.001; 0.619, p = 0.385). Within the HBeAg-negative patient subgroup, the AUC for IL-34 surpassed that of FIB-4 (p = 0.009) and APRI (p = 0.045). Conclusion: Serum IL-34 has the potential to be a straightforward and practical biomarker that demonstrates superior performance to serum CSF-1 in the diagnosis of significant liver fibrosis in CHB patients, especially within the HBeAg-negative patients.
Article
Background: Hepatitis B Virus (HBV) is one of the leading causes of infectious disease in the global population, and its prevalence has been increasing globally. Human HBV infection is complex, involving both innate and adaptive immune systems. Cytokines play a role in both physiologic and pathologic processes in the liver. This study was designed to screen serum levels using an enzyme linked immunosorbent assay (ELISA) and genetic variants in the TNF-α and IL6 genes using polymerase chain reactions (PCR). The aim of this study was to screen the serum levels and genotype levels with TNF-α (C-308 T/G-308A) and IL-6 (G-174 C) genes in HBV patients and control subjects. Methods: In this study, we have selected 50 HBV patients and 40 control subjects from Saudi Population. Patient serum samples was used for measuring the serum levels and PCR analysis using RFLP analysis. Prior to this, HBV patients were confirmed with PCR analysis followed by Sanger sequencing analysis. Results: The current study results confirmed positive association in serum levels (p < 0.05) and negative association with both genotype and allele frequencies in TNF-α (C-308 T) and IL-6 (G-174 C) genes among HBV patients and controls (p > 0.05). Positive associations between blood levels of TNF-α and IL-6 were confirmed, while negative associations were found between PCR investigations involving the TNF-α (G-308A) and IL-6 (G-174 C) genes with the HBV prevalence in the Saudi population. Conclusion: This study confirmed serum levels are strongly associated with HBV patients in the Saudi population. However, PCR studies showed the negative association with the couple of variants selected for this study.
Article
Full-text available
Background: Immune cells and molecules play a vital role in initiating, maintaining, regulating immunological homeostasis and inflammation in many pathological and physiological processes; however, the changes on expressions and functions of these cells and molecules in hepatitis B virus (HBV) infection have not been elucidated well. Objectives: The current study aimed to determine the expression pattern of different cytokines, chemokines, immune cells in HBV infection and their association with disease progression. Patients and Methods: Sixty-nine patients with chronic HBV infection were enrolled. Five immune cell subsets and 46 cytokines and chemokines were analyzed by flow cytometry and Luminex 200. Results: In comparison to healthy individuals and asymptomatic HBV carriers, expression of CXCL9, CXCL10, CXCL11, and IL-10 were elevated in patients with chronic active HBV and had positive correlation with ALT levels. In contrast, G-CSF, MCP-3, and IFN-γ levels were significantly decreased in patients with chronic active HBV infection in contrast to carriers and healthy individuals; however, these down regulations did not show any correlation with either virological findings or liver inflammation. Although the proportion of CD4+ CD25 high regulatory T cells (Tregs) was higher in patients with HBV infection than in healthy controls, no correlations were found between Tregs and other cytokines or chemokines. Conclusions: CXCR3-associated chemokines might contribute to liver inflammation in chronic hepatitis B, while MCP-3 and G-CSF were inhibited by HBV infection. Host immune response was suppressed as manifested by an increase in CD4+ CD25high Tregs and IL-10 as well as a decrease in IFN-γ. Exploiting the expression pattern of cytokine and chemokine may help to develop a better understanding of chronic HBV infection pathogenesis.
Article
Full-text available
Abstract The major shortcoming of nucleos(t)ide analogue therapy for chronic hepatitis B (CHB) is the frequent requirement for indefinite therapy. Withdrawal of treatment can result in viral rebound with an alanine aminotransferase (ALT) flare leading to hepatic decompensation, while in others this can lead to hepatitis B e antigen (HBeAg) seroconversion. The aim of the study was to identify host immune profiles associated with these different outcomes. Eighteen HBeAg(+) patients, enrolled on a phase III trial with the nucleoside analogue adefovir dipivoxil, were followed for up to 128 weeks. For the first 48 weeks, all patients received continuous therapy. Subsequently, patients experienced cycles of treatment interruptions due to random drug/placebo misallocations. Host immune profiles were characterized by measuring a panel of serum cytokines before, during, and after each cycle of treatment withdrawal. Virus-specific T-cell responses were also determined at these time points in a subset of patients to elucidate the mechanisms utilized to control hepatitis B virus (HBV) replication post-treatment. Significantly, elevated levels of IFN-γ, IP-10, and IL-2 on-treatment were associated with HBeAg seroconversion after treatment withdrawal. In these patients, treatment interruption induced further increases in serum IFN-γ levels and marked increases in virus-specific T-cells producing IFN-γ, but minimal alterations in viremia and ALT. In HBeAg(+) patients with low on-treatment levels of serum IFN-γ, the interruption of therapy induced significant elevations in HBV-DNA, ALT, IP-10, and increases in virus-specific T-cells producing IL-10. Evaluating on-treatment serum cytokines in concert with virologic and clinical parameters may help to identify CHB patients who can successfully discontinue nucleos(t)ide analogue therapy.
Article
Full-text available
Introduction: Hepatitis B virus (HBV) infection is a global health problem. Peginterferon α (PEG-IFN), which includes PEG-IFN α-2a (Pegasys) and PEG-IFN α-2b (Peg-Intron), can be used to treat patients with chronic hepatitis B (CHB) infection. A finite duration of PEG-IFN therapy may lead to long-term viral suppression. Clinically, it is important to identify super-responders and null-responders to PEG-IFN due to its substantial side effects. Areas covered: From the literature review, it is known that PEG-IFN is more effective for hepatitis B e antigen (HBeAg)-positive patients who have high pre-treatment alanine aminotransferase level, lower HBV DNA level and genotype A (vs genotype D), as well as those with more favourable viral predictors, such as precore stop codon or basal core promoter mutants infections in Asian patients and wild-type virus in Caucasian patients. For HBeAg-positive patients and HBeAg-negative patients with genotype D infection, PEG-IFN therapy could be terminated early at week 12 or 24 in primary non-responders defined by the Hepatitis B surface antigen stopping rules. With regard to host factors, single nucleotide polymorphisms of IL28B do not seem to affect the treatment outcomes in Asian patients, but its role in Caucasian patients remains disputed. Expert opinion: Most of the known predictors need validation by large prospective trials. In addition, we need to identify more baseline predictors for super-responders in order to achieve personalised PEG-IFN treatment for CHB.
Article
A cytokine synthesis inhibitory factor (CSIF) is secreted by Th2 clones in response to Con A or antigen stimulation, but is absent in supernatants from Con A-induced Th1 clones. CSIF can inhibit the production of IL-2, IL-3, lymphotoxin (LT)/TNF, IFN-gamma, and granulocyte-macrophage CSF (GM-CSF) by Th1 cells responding to antigen and APC, but Th2 cytokine synthesis is not significantly affected. Transforming growth factor beta (TGF-beta) also inhibits IFN-gamma production, although less effectively than CSIF, whereas IL-2 and IL-4 partially antagonize the activity of CSIF. CSIF inhibition of cytokine synthesis is not complete, since early cytokine synthesis (before 8 h) is not significantly affected, whereas later synthesis is strongly inhibited. In the presence of CSIF, IFN-gamma mRNA levels are reduced slightly at 8, and strongly at 12 h after stimulation. Inhibition of cytokine expression by CSIF is not due to a general reduction in Th1 cell viability, since actin mRNA levels were not reduced, and proliferation of antigen-stimulated cells in response to IL-2, was unaffected. Biochemical characterization, mAbs, and recombinant or purified cytokines showed that CSIF is distinct from IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IFN-gamma, GM-CSF, TGF-beta, TNF, LT, and P40. The potential role of CSIF in crossregulation of Th1 and Th2 responses is discussed.
Article
Hepatitis B virus (HBV) infection is still a public health problem worldwide, being endemic in some parts of the world. It can lead to serious liver diseases such as chronic hepatitis, cirrhosis, and hepatocellular cancer. The differences in host immune response can be one of the reasons for the various clinical presentations of HBV infection. Polymorphisms of genes encoding the proinflammatory and antiinflammatory cytokines, which are responsible for regulation of the immune response, can affect the clinical presentation of the infection. Particularly, the polymorphisms of the genes encoding cytokines such as interleukin (IL)-1, IL-6, IL-8, IL-10, IL-18, IL-28B, interferon-γ, tumor necrosis factor-α, tumor growth factor-β1, and regulatory molecules like vitamin D receptor and chemokine receptor 5 can be responsible for different clinical presentations of HBV infections. The genomic information about cytokines and other mediators can be important for determining high-risk people for developing chronic hepatitis or hepatocellular cancer and may be used to plan treatment and preventive approaches for these people. In this review, the current knowledge in the literature on the association between cytokine/regulatory molecule gene polymorphisms and clinical course of chronic HBV infection is summarized, and the clinical implementations and future prospects regarding this knowledge are discussed.
Article
Hepatitis B is one of the most prevalent infectious diseases and is induced by hepatitis B virus (HBV). The chronic, asymptomatic, and occult forms of hepatitis B are long-term infections that can lead to various hepatic cancers and cirrhosis in the carrier. IL-12 is one of the main cytokines involved in inducing appropriate immune responses against viral infections, especially HBV. Therefore, the aim of the present review was to address the most recent information within the database regarding the status and association between IL-12 and hepatitis B infection and its complications including cirrhosis and hepatocellular carcinoma. The data presented was collected by searching the following keywords in the Pubmed and Scopous databases: Hepatitis B, occult HBV infection, chronic HBV infection, asymptomathic HBV infection, acute HBV infection, fulminant HBV infection, IL-12, and all the papers regarding the relation between IL-12 and hepatitis B were used. These data were presented in the current review article. Results showed that IL-12 plays important roles in Hepatitis B infection and patients infected with the long-term form of hepatitis B are unable to produce sufficient amount of this cytokine. Our research provided mechanistic insights into the immunoprotective roles of IL-12 and proposed that it can be considered as an important molecule for immunotherapy of HBV infected infection.
Article
Hepatitis B (HBV) infection is a major cause of chronic liver diseases, and the polymorphisms of cytokine genes may affect the progression of HBV-related hepatitis. The aim of this study was to examine the association of cytokine polymorphisms with the susceptibility to HBV-related chronicity. Specifically, a LIFECODES Cytokine SNP Typing kit was used to investigate 22 cytokine single nucleotide polymorphisms (SNPs) from 14 cytokine and cytokine receptor genes with the aim of analyzing the role of Th1 and Th2 genotype combination. This population-based case-control association study included 131 chronic HBV patients and a control group of 142 healthy donors. When the combination of Th1 and Th2 genotypes was analyzed for the genetic risk factor for chronic hepatitis B, we did not observe any significant association. A non-significant association betweenTh1 and Th2 and this risk factor could have resulted from the limitation ofour small sample size. When the results from each genotype were separately analyzed, the frequencies of the heterozygous CA (-592) and CT (-819) genotype of IL-10 gene-promoter polymorphisms were significantly higher in chronic HBV patients than that in healthy controls (OR=1.76, 9%CI =1.03-3.01, p =0.028; OR=1.79, 95%CI =1.04-3.06, p =0.024, respectively). Interestingly, the TCC (-1098/-590/-33) haplotype frequency of IL-4 showed a positive association with chronic hepatitis B as a protective haplotype (OR =0.53, 95%CI =0.32-0.85, p =0.005). Conclusion: These preliminary results suggest that polymorphisms in some cytokine genes, particularly the Th2 cytokine, influence persistence of HBV infection.