Editorial Open Access
Copyright ©2008 The WJG Press and Baishideng. All rights reserved.
World J Gastroenterol. Dec 21, 2008; 14(47): 7149-7159
Published online Dec 21, 2008. doi: 10.3748/wjg.14.7149
Role of chemokines and their receptors in viral persistence and liver damage during chronic hepatitis C virus infection
Juan R Larrubia, Eduardo Sanz-de-Villalobos, Liver Research Unit, Department of Gastroenterology, Guadalajara University Hospital, University of Alcalá, Donante de Sangre st. 19002 Guadalajara, Spain
Selma Benito-Martínez, Miryam Calvino, Trinidad Parra-Cid, Liver Research Unit, Guadalajara University Hospital, University of Alcalá, Donante de Sangre st. 19002 Guadalajara, Spain
Author contributions: Larrubia JR and Benito-Martínez S contributed equally towards the conception and design of the review; Larrubia JR wrote and revised the manuscript; Calvino M, Sanz-de-Villalobos E, and Parra-Cid T contributed equally to the supportive work and supervision.
Supported by Grants from “Fiscam” J.C.C.M (Ayuda para proyectos de investigación en salud; PI-2007/32), “Asociación Castellana de Aparato Digestivo” (Beca ACAD; ACAD/06) and “Fundación de Investigación Médica Mutua Madrileña” (Beca Ayudas a la Investigación FMMM; 2548/2008), Spain; Selma Benito-Martínez was supported by a research grant from “Fiscam” J.C.C.M (“Perfeccionamiento y movilidad de investigadores”; MOV-2007_JI/18), Spain; Miryam Calvino was supported by a research grant from “Instituto de Salud Carlos III” (Contrato de apoyo a la investigación en el SNS’’; CA07/00157), Spain
Correspondence to: Juan R Larrubia, MD, MSc, PhD, Department of Gastroenterology, Guadalajara University Hospital, University of Alcalá, Donante de Sangre st. 19002 Guadalajara, Spain. jlarrubiam@meditex.es
Telephone: +34 -949-209200 Fax: +34-949-209259
Received: August 7, 2008
Revised: November 26, 2008
Accepted: December 2, 2008
Published online: December 21, 2008

Abstract

Chemokines produced in the liver during hepatitis C virus (HCV) infection induce migration of activated T cells from the periphery to infected parenchyma. The milieu of chemokines secreted by infected hepatocytes is predominantly associated with the T-helper/T-cytotoxic type-1 cell (Th1/Tc1) response. These chemokines consist of CCL3 (macrophage inflammatory protein-1α; MIP-1α), CCL4 (MIP-1β), CCL5 (regulated on activation normal T cell expressed and secreted; RANTES), CXCL10 (interferon-γ−inducible protein-10; IP-10), CXCL11 (interferon-inducible T-cell α chemoattractant; I-TAC), and CXCL9 (monokine induced by interferon γ; Mig) and they recruit T cells expressing either CCR5 or CXCR3 chemokine receptors. Intrahepatic and peripheral blood levels of these chemokines are increased during chronic hepatitis C. The interaction between chemokines and their receptors is essential in recruiting HCV-specific T cells to control the infection. When the adaptive immune response fails in this task, non-specific T cells without the capacity to control the infection are also recruited to the liver, and these are ultimately responsible for the persistent hepatic damage. The modulation of chemokine receptor expression and chemokine secretion could be a viral escape mechanism to avoid specific T cell migration to the liver during the early phase of infection, and to maintain liver viability during the chronic phase, by impairing non-specific T cell migration. Some chemokines and their receptors correlate with liver damage, and CXCL10 (IP-10) and CXCR3 levels have shown a clinical utility as predictors of treatment response outcome. The regulation of chemokines and their receptors could be a future potential therapeutic target to decrease liver inflammation and to increase specific T cell migration to the infected liver.

Key Words: Chemokines, Chemokine receptors, Hepatitis C virus, Viral hepatitis pathogenesis, Persistent infection, Viral escape mechanism



INTRODUCTION
Table 1 Chemokines and their receptors.
Chemokine receptorChemokine ligandsTarget cells
Subfamily CC
CCR1CCL3, CCL5, CCL7, CCL14T cells, monocytes, basophils and eosinophils
CCR2CCL2, CCL8, CC7, CCL13, CCL16Memory T cells, monocytes and dendritic cells
CCR3CCL11, CCL13, CCL7, CCL5, CCL8, CCL13Eosinophils, basophils, mast cells, T helper 2 cells and platelets
CCR4CCL17, CCL22T helper 2 cells, dendritic cells, basophils, macrophages and platelets
CCR5CCL3, CCL4, CCL5, CCL11, CCL14, CCL16T cells, monocytes
CCR6CCL20T cells, B cells and dendritic cells
CCR7CCL19, CCL21T cells and dendritic cells
CCR9CCL25T cells, plasma cells
CCR10CCL27, CCL28T cells
Subfamily CXC
CXCR1CXCL8, CXCL6Neutrophils and monocytes
CXCR2CXCL8, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6Neutrophils, monocytes and vascular endothelial cells
CXCR3-ACXCL9, CXCL10, CXCL11T helper 1 cells, mast cells and mesangial cells
CXCR3-BCXCL4, CXCL9, CXCL10, CXCL11Neoplastic cells and vascular endothelial cells
CXCR4CXCL12Expressed in multiple cells
CXCR5CXCL13B cells and T helper cells
CXCR6CXCL16CD8+ cells, natural killer cells and memory CD4+ cells
Subfamily CX3C
CX3CR1CX3CL1Macrophages and smooth muscle cells
Subfamily XC
XCR1XCL1, XCL2T cells and natural killer cells
Figure 1
Figure 1 Innate and adaptive immune response. Importance of specific cytotoxic T cell response. In a non-cytopathic viral infection the development of a vigorous specific cytotoxic T lymphocyte (CTL) response is essential. Professional antigen presenting cells (APC) take up viral antigens and migrate from infected parenchyma to the lymph nodes to prime naive specific CTLs. These cells express the chemokine receptor CCR7 to reach the lymph nodes, attracted by CCL1 and CCL2 chemokines. After priming, specific CTLs lose CCR7 expression and up-regulate CCR5 and CXCR3 chemokine receptors, then migrate to the hepatic parenchyma to develop their effector function, attracted by the chemokines produced in the liver. During the early phase of infection, innate immunity is the first barrier for fighting against the virus. The cellular innate response is also recruited in the infected parenchyma by the interaction between chemokines and their receptors.
Figure 2
Figure 2 Different migration potential between resolved and persistent HCV infection. In a resolved infection HCV-specific cytotoxic T lymphocytes (CTL) migrate rapidly to the infected liver, attracted by the chemokines produced by hepatocytes, succeeding in controlling HCV infection without liver damage. In persistent infection the migration of T cells to the liver may be impaired. High chemokine levels during infection may induce chemokine receptor down-regulation on T cells by molecule internalization. This mechanism could impair the migration of specific T cells during primary infection, which could inhibit HCV clearance. During the chronic phase, the persistent presence of HCV in the liver could keep a long-standing chemokine production, which could attract non-specific mononuclear cells to the liver, and be responsible for chronic liver damage. The relative migration impairment due to chemokine receptor down-regulation on non-specific T cells could modulate inflammatory infiltration of the liver, inducing chronic low-grade liver damage, which could favour host and virus long-term survival.
Figure 3
Figure 3 Chemokines and chemokine receptors related to chronic hepatitis C pathogenesis. In chronic hepatitis C, intrahepatic chemokines recruit Th1/Tc1 cells expressing CCR5/CXCR3 chemokine receptors. The X symbol indicates that chemokines and cytokines associated with a Th2/Tc2 response are not primarily produced in the HCV infected liver.
Figure 4
Figure 4 Adequate chemotaxis is necessary to control a viral infection. Specific cytotoxic T cells with active in vitro effector functionality are not able to control a viral infection if they do not express the appropriate chemokine receptor to reach the infected parenchyma. In a murine model of influenza virus infection, specific CCR5-expressing T cells were able to clear the virus while administration of specific CCR3-expressing T cells induced mouse death. CTLs: cytotoxic T lymphocytes.
Figure 5
Figure 5 Intrahepatic chemoattraction of non-specific T cells causes liver damage. In a murine model of graft versus host disease the liver is infiltrated by CCR5-expressing T cells causing liver failure. This process can be blocked by using anti-CCR5 monoclonal antibodies.
Figure 6
Figure 6 Intrahepatic enrichment of CCR5 and CXCR3 expressing T cells. FACScan® dot-plots of liver infiltrating (LIL), and peripheral blood (PBL) lymphocytes from a patient with chronic hepatitis C stained with labelled antibodies against CD8, CCR5 and CXCR3. The intrahepatic frequency of CCR5/CXCR3-expressing CD8+ cells is higher than in the peripheral blood.
Figure 7
Figure 7 Correlation between liver inflammation and frequency of intrahepatic CCR5 and CXCR3 expressing T cells in chronic hepatitis C. A: Representative photomicrographs of liver immunostaining for CD8 from two chronic hepatitis C patients with different grades of inflammation. CD8 cells are stained in dark by the immunoperoxidase technique. Original magnification 400X. A patient with lobular activity (LA) 1 and porto-periportal activity (PPA) 1 showed little staining while a patient with LA 3 and PPA 3 presented intense CD8 staining; B: FACScan® dot-plots of intrahepatic lymphocytes from these two patients after staining with CD8-Pe and with either CCR5-Cy5 or CXCR3-FITC mAbs. In the upper right quadrant are represented the double positive cells. The frequency of CCR5/CXCR3 expressing CD8+ cells is higher in the patient with the higher histological liver inflammation.

The hepatitis C virus (HCV) is a hepatotropic non cytopathic virus very efficient in evading the host immune response. Hepatitis C infection is a major cause of chronic liver disease worldwide, affecting at least 170 000 000 people[1]. Approximately three quarters of infected subjects develop a chronic infection, but only one third progress to cirrhosis, hepatocellular carcinoma and liver failure without treatment[2]. Nowadays around 50% of patients treated with pegylated-interferon plus ribavirin clear the virus[3,4]. To control HCV infection an adequate specific T cell response is necessary[5-7], with T cells that are able to migrate to the infected site to develop their effector functions (Figure 1). Nevertheless, specific T cells fail to remove the virus in the majority of patients[7,8], because often a non-specific T cell population is recruited to the infection site, and these cells are presumably responsible for the chronic damage[9] (Figure 2).

In both scenarios the attraction of leukocytes to the liver is controlled by chemokines, which are chemotactic cytokines secreted by infected cells and interact with their receptors expressed on the recruited leukocytes[10] (Figure 3). In an experimental model of influenza virus infection, the importance of appropriate chemotaxis to control viral infection was shown. In this model, cytotoxic specific CD8+ cells expressing inadequate chemokine receptors were not able to reach the infected site and clear the virus, while specific cytotoxic T cells expressing the correct chemokine receptors controlled the infection without tissue damage[11] (Figure 4). On the other hand, it has also been demonstrated that chemokines and their receptors can be involved in liver damage. In an animal model study it was reported that a massive hepatic infiltration by non-specific T cells, expressing chemokine receptors associated with the type-1 response, can cause acute liver failure[12] (Figure 5). Therefore, chemokines and their receptors are associated with viral control but are also associated with immune-mediated liver inflammation. Moreover, in a hepatotropic viral infection in humans, a huge intrahepatic non-specific mononuclear infiltrate during viral persistence was noticed, while this was not present in subjects with viral control[13] (Figure 2). In this last study, the intrahepatic chemoattraction of non-specific T cells perpetuated the liver damage. Consequently, also in humans, chemokines and their receptors develop an important role in viral clearance and in the development of chronic tissue inflammation. Obviously, the modulation of these pathways is important for generating an efficient immune response, and for participating in the inflammatory process during the chronic infection phase, but pathway modulation could also be a viral strategy used by HCV to escape from immune control[14]. This review introduces the advances obtained in the last decade on the role of chemokines and their receptors in chronic hepatitis C pathogenesis.

STRUCTURE AND FUNCTION OF CHEMOKINES AND THEIR RECEPTORS

Chemokines are small heparin-binding proteins that direct the movement of mononuclear cells through the body to contribute to the development of an adaptive immune response and to the pathogenesis of inflammation. These molecules also play a role in angiogenesis, haematopoiesis, lymphoid organ development, wound healing and regulation of embryonic development. These proteins are 8-10 kDa in size with 20%-70% amino acid sequence homology and are secreted by resident cells at the inflammatory site[10]. Around 50 chemokines have been described and are subdivided into four families according to the position of the two N-terminal cysteine residues: CXC, CC, XC and CX3C[15,16,17] (Table 1). The CXC family has also been subdivided into two groups depending on the presence of the ELR motif (Glu-Leu-Arg). A systematic nomenclature has been adopted in the past few years for chemokines and their receptors[18,19]. Chemokines induce cell migration and activation by binding to specific G-protein-coupled cell-surface receptors on target cells, called chemokine receptors[10] (Table 1). Receptor triggering leads to a cascade of cellular activation, including the generation of inositol triphosphate, the release of intracellular calcium, and the activation of protein kinase C[20]. Chemokine receptor signalling also activates small guanosine triphosphate binding proteins of the Ras and Rho families[21]. Rho proteins are involved in cell motility through regulation of actin-dependent processes such as membrane ruffling, pseudopod formation, and assembly of focal adhesion complexes[15]. All these mechanisms propel cells in the appropriate direction. In humans different chemokine receptors subdivided into four families have been described: XC, CXC, CC and CX3C chemokine receptors. These receptors are expressed on different types of leukocytes, and some are constitutively expressed while others are induced, depending on the degree of leukocyte activation and differentiation[15]. Polarisation of chemokine receptor expression on T cells depending on the cytokine production profile has been demonstrated[22]. Chemokine receptors, such as CCR5 and CXCR3, are associated with the type-1 response, while CCR3, CCR4 and CCR8 are associated with the type-2 response[23,24,25,26]. Due to the preferential Th1/Tc1 response of liver infiltrating T cells during chronic hepatitis C[27,28], this review focuses on these two chemokine receptors, which bind chemokines from the non-ELR-CXC and CC subfamilies. The ligands for CXCR3 are interferon (IFN)-γ-inducible protein 10 (IP-10, CXCL10), monokine induced by IFN-γ (Mig, CXCL9), and IFN-inducible T-cell α chemoattractant (I-TAC, CXCL11). CXCR3 is expressed on activated T cells and natural killer cells[29]. The CCR5 ligands comprise regulated upon activation, normal T-cell expressed and secreted (RANTES, CCL5), macrophage inflammatory proteins 1α (MIP 1α, CCL3) and 1β (MIP 1β, CCL4). CCR5 is expressed predominantly on activated and memory T cells. Hereafter the systematic nomenclature for chemokines will be used.

CHEMOKINE SECRETION AND CHEMOKINE RECEPTOR EXPRESSION IN CHRONIC HEPATITIS C

The migration of lymphocytes to the liver is a complex process involving adhesion, rolling, triggering, and transendothelial migration. Chemokines and their receptors play an essential role in this multistep pathway[30,31]. In chronic hepatitis C, the expression of different chemokines in the liver has been described. CXCL10 is increased in the liver and peripheral blood during chronic hepatitis C[32,33,34,35]. This molecule is produced by hepatocytes and sinusoidal endothelial cells[33,34]. CXCL9 and CXCL11 are also increased in the serum and liver of subjects with chronic hepatitis C[33,36]. CXCL9 is detected primarily on sinusoidal endothelial cells, while CXCL11 is produced mainly by hepatocytes[33,37]. The intrahepatic expression of CCL5 is also elevated in chronic hepatitis C and it is produced by hepatocytes, sinusoidal endothelial cells and biliary epithelium[37]. Finally, several studies have reported an increased level of CCL3 and CCL4 either in the liver or in serum. These molecules are detected on endothelial cells, on some hepatocytes and on biliary epithelial cells[33,35,38]. The expression of all these chemokines in the liver can be induced directly by HCV. Previous reports have shown a high hepatocyte synthesis of CXCL10, CXCL9 and CCL5, induced by some HCV proteins such as NS5A and core[39], although a recent in vitro study suggests that HCV proteins could also decrease the expression of CCL5 and CXCL10 genes[40]. All these chemokines recruit T cells with a Th1/Tc1 phenotype expressing specific chemokine receptors such as CCR5 and CXCR3[24]. The non-ELR-CXC chemokine attracts CXCR3 expressing T cells while CC chemokine attracts CCR5 expressing T cells to the liver. Consequently, in chronic hepatitis C, an intrahepatic enrichment of CCR5 and CXCR3 expressing T cells, located in the hepatic lobule and portal tracts, has been shown while these populations are very infrequent in uninfected subjects[33,35,37] (Figure 6).

CORRELATION BETWEEN LIVER INFLAMMATION AND CHEMOKINE/CHEMOKINE RECEPTOR

Persistent HCV infection is characterised by a non-specific inflammatory infiltrate in the liver, mainly composed of CD8+ cells[41,42], and responsible for liver damage[9]. These cells are attracted by the interaction between the intrahepatic secreted chemokines and the chemokine receptors expressed on T cells. Previous reports have shown a correlation between liver inflammation and liver infiltrating CXCR3/CCR5 expressing T cells[35,37] (Figure 7). The frequency of occurrence of these cells was positively correlated with portal and lobular inflammation but not with liver fibrosis. These data suggest that CCR5 and CXCR3 could play an important role in chronic liver damage by means of recruitment of inflammatory T cells into the liver. Several previous studies have also shown a correlation between liver inflammation and chemokine levels. Intrahepatic CXCL10 mRNA levels are associated with intralobular inflammation[43]. Similarly, CXCL9 and CXCL11 correlate with the grade of liver inflammation[37,44]. Furthermore, CC chemokines are also correlated with intrahepatic inflammatory activity[45]. Clearly, intrahepatic CCL5-positive cells correlate with inflammatory activity but not with liver fibrosis[37]. The absence of correlation between liver fibrosis and liver infiltrating CXCR3/CCR5 expressing T cells and their ligands could be due to a statistical beta error, but also could suggest that it is necessary for something other than liver inflammation to occur in order to develop fibrosis, such as genetic and non-genetic factors[46]. Nevertheless, a recent study, examining a large sample of chronic hepatitis C patients, showed a positive correlation between CXCL9, CXCL10 and CXCL11 intrahepatic levels and the grade of fibrosis[47], indicating that persistent liver inflammation produced by the mononuclear cells attracted by these chemokines could finally induce the activation of a liver fibrosis cascade. Bearing in mind all the previous data, it is possible to speculate that chemokines are secreted in the infected liver to attract an adaptive immune response able to clear the virus. Unfortunately, when the specific response fails these chemokines also attract non-specific T cells, which are not able to remove the virus but produce liver inflammation (Figure 2). Therefore, as chemokines are nonspecific chemoattractants, the intrahepatic inflammatory infiltrate produced during chronic infection is mainly non-HCV-specific and consequently unable to eliminate HCV. It is, however, able to produce cytokines capable of initiating and perpetuating hepatic fibrogenesis[48]. The efficiency of this mechanism could play a role in determining why chronically infected individuals either do or do not progress to fibrosis. Certain polymorphisms in key chemokines known to be up-regulated in chronic HCV, such as CCL5, have been identified as correlates with the development of fibrosis[49].

MODULATION OF CHEMOKINE/CHEMOKINE RECEPTOR PATHWAY AS A VIRAL ESCAPE MECHANISM

HCV is usually able to evade the immune system efficiently. Several HCV escape mechanisms have been previously described, such as selection of escape mutations, induction of specific T cell anergy or resistance to the effects of α-interferon[5,50]. One hypothetical mechanism for HCV to survive could be to reduce hepatic chemotaxis of T lymphocytes to impair infection control during primary infection and also to decrease tissue damage during the chronic phase. The natural history of chronic hepatitis C means it can take up to three decades to develop liver cirrhosis[51]. This in turn means that the immunologically-mediated liver damage must be continuous but very light. For HCV it is essential to extend host survival as much as possible to assure its own viability. Considering these facts, it could be important for HCV to impair the expression of chemokine receptors associated with the type-1 response to improve its survival ability.

One mechanism to achieve this could be to reduce T cell migration into the liver through impairment of CCR5 and CXCR3 expression. To maximise the ability of the immune system to control the infection, a high frequency of HCV-specific CCR5+/CXCR3+ T cells should be expected during primary infection. Soon after HCV infection, prominent CD8+ cell responses are repeatedly observed and transient up-regulation of CCR5 and CXCR3 expression is seen[52,53]. During chronic hepatitis C, however, T cells show reduced surface expression of chemokine receptors associated with the Th1/Tc1 response while an intracellular increase of these molecules has been described[54]. This finding suggests the occurrence of HCV-mediated chemokine receptor internalization in chronically infected subjects. Moreover, a high serum concentration of CCL3 and CXCL10, associated with a normal or reduced peripheral frequency of CCR5- and CXCR3-positive T cells during chronic hepatitis C, has been also described[35,54]. During chronic HCV infection, the observed absence of an increase in the number of peripheral CCR5/CXCR3-positive T cells could be due to either an intrahepatic sequestration of CCR5/CXCR3 expressing T cells, caused by CCL3/CXCL10 attraction, or to a down-regulation of these chemokine receptors produced by the high serum concentration of their ligands.

It has been shown that GB virus C, a close relative of HCV[55], can reduce CCR5 expression on T cells by inducing CCL5 release[56,57]. This mechanism impairs the ability of HIV to infect CD4 T cells, thus extending host survival[57,58]. Another study into HCV infection described CCR5 down-regulation on CD8+ cells by receptor internalization[54]. It has been shown that the HCV-E2 protein, after binding to CD81[59], induces CCL5 secretion by CD8+ cells and the ensuing interaction between CCL5 and CCR5 is responsible for CCR5 down-regulation on these cells[60]. All these data suggest that HCV could modulate chemokine receptors associated with the Tc1 response to achieve a survival advantage. This mechanism could decrease HCV-specific T cell migration during acute infection, avoiding viral control, and could also impair non-specific T cell migration during persistent infection, modulating liver inflammation and fibrosis which could extend host and viral survival. If HCV is able to interfere with CCR5/CXCR3 expression, an increase in T cells expressing these chemokine receptors after viral load drop due to anti-viral treatment should be expected, together with a CXCL10/CCL3 decrease. To address this significant issue, a longitudinal analysis of CCR5/CXCR3 expressing CD8+ cells and CXCL10/CCL3 levels during treatment was performed[35]. In the majority of treated patients in this study, an increase in CCR5/CXCR3 expressing CD8+ cells was demonstrated. This finding was associated with a significant decrease in CXCL10 and CCL3 serum levels after 24 wk of treatment. A likely explanation for these data is that HCV control during treatment could decrease CXCL10/CCL3 release, allowing CCR5/CXCR3 up-regulation on peripheral CD8+ cells. Actually, in this study a significant positive correlation between HCV viral load and CCL3 and an almost significant correlation with CXCL10 was shown. In summary, all these data taken together suggest that HCV could down-regulate CCR5 and CXCR3 expression on T cells by means of the secretion induction of their chemokine agonists. This strategy could favour HCV escape from immune control and could also decrease long-standing liver inflammation, allowing extended host and virus survival.

CHEMOKINE/CHEMOKINE RECEPTOR LEVELS AS TREATMENT RESPONSE OUTCOME PROGNOSTIC TOOL

As previously commented, Tc1/Th1 associated chemokines and their ligands can be modulated by HCV infection to impair the immune response. Therefore, the levels of these molecules may also influence treatment-mediated viral clearance. Previous studies have shown how baseline CXCL10 serum concentration is associated with the outcome of antiviral therapy in monoinfected[61-64] patients and in patients co-infected with HIV[65]. Elevated pre-treatment CXCL10 levels correlate with non-response to current therapy. Moreover, it was shown that the increase in CXCR3 expressing CD8+ cells during treatment is associated with SVR[35]. This suggests that for HCV, it is important to modulate the expression of this receptor not only to maintain liver viability but also to escape from immunological control. In addition, in this last study, a faster reduction in CXCL10 serum concentration was suggested in responders than in non-responders during the first 12 wk of treatment. Therefore, high CXCL10 may decrease the response of CXCR3 expressing T cells and have a negative influence on treatment outcome. Patients with either low base-line or rapid decrease of CXCL10 levels could promptly restore the response of CXCR3 expressing T cells which could assist in viral control. On the other hand, the absence of an increase in CXCR3 expressing CD8+ cells after 24 wk of treatment is associated with a 100% negative predictive value of SVR[35]. This information may be clinically important in predicting non-response and allowing the termination of treatment in those patients with no increase in the frequency of CXCR3 expressing CD8+ cells after 24 wk of treatment. Nevertheless, with these data it is impossible to completely rule out the possibility that the observed change in chemokine patterns during treatment was an epiphenomenon due to a direct interferon effect[66]. This aspect could be clarified by studies based on patients treated with protease inhibitors. Finally, CCL20 and CXCL9 have also been suggested as tools to predict treatment outcome[61,67]. All these studies defined to predict treatment response provide preliminary information only since they are based on a very small number of patients. Therefore, these data should be re-confirmed by larger multivariate studies before applying these prognostic variables to daily clinical work.

ROLE OF CHEMOKINE/CHEMOKINE RECEPTOR POLYMORPHISMS

Several chemokine and chemokine receptor polymorphisms have been associated with different HCV infection outcomes. A CXCL11 polymorphism, defined by a 5-bp deletion in the CXCL11 promoter, has been associated with an increased risk of chronic HCV infection[68]. The distribution frequency of the allele was found to be significantly increased in a chronically HCV infected population compared to healthy controls. This deletion variant significantly reduced the transcriptional activity of the CXCL11 promoter in vitro in the presence of replicating HCV, which impaired T cell migration in vivo. Moreover, a CCR5 frameshift mutation, called CCR5-Δ32 that abrogates CCR5 expression also favoured HCV infection[69]. In this study an association between this homozygous mutation and an increased prevalence of HCV infection, as well as an increase in viral load, was documented. On the other hand, this mutation has also been associated with reduced liver inflammation[49,70,71]. These data suggest that the specific and non-specific T cell migration impairment due to the absence of CCR5 expression could favour HCV persistence and could also decrease liver damage. Similarly, a CCL5 deletion mutation which produces higher expression of CCL5 is associated with a lower grade of liver inflammation and fibrosis[70,71]. In this case the CCL5 over-expression may lead to CCR5 internalization and subsequent impairment of T cell migration. These mutations, which reduce chemokine receptor expression, are interesting proofs of the role of these molecules in the development of intrahepatic inflammation during chronic hepatitis C.

CHEMOKINE RECEPTORS AS A POTENTIAL THERAPEUTIC TARGET

Human monoclonal antibodies against CCR5, CXCR3 and their ligands have been used to treat different inflammatory and infectious diseases in humans and in animal models. CCR5 monoclonal antibodies have been shown to be effective in avoiding T cell infection by CCR5-tropic HIV-1[72,73,74], and also in decreasing tissue inflammation in some animal models[75]. CXCL10 blocking is a successful treatment for experimental colitis[76,77]. Anti-CXCR3 monoclonal antibodies display an anti-inflammatory effect in an animal model of arthritis[78] and could also be a therapeutic target in inflammatory bowel disease[79]. These molecules have not been yet tested in chronic hepatitis C, due to the absence of an adequate animal model for proving their efficiency and safety. In the near future, an HCV permissive mouse model reconstituted with a human immune system will allow us to study mechanisms of chemokine/chemokine receptor immunopathogenesis[80]. Nevertheless, in a murine model of liver failure the administration of anti-CCR5 monoclonal antibodies has been shown to suppress intrahepatic liver inflammation, allowing mouse survival[12] (Figure 5). At least theoretically, all these previous data suggest that antibodies able to block the interaction between CCR5/CXCR3 and their ligands could decrease chronic liver inflammation in HCV infected subjects unable to clear the virus. Clearly, if the migration of non-specific T cells to the infected liver is impaired, liver damage will be reduced since HCV is not directly cytopathic. This strategy could be explored in patients without a sustained virologic response after current standard treatment. These future drugs could reduce liver fibrosis progression until new effective anti-HCV treatments are available. Obviously, the lessons learnt from HIV about the safety of these drugs should be the milestone for starting clinical trials in non-responder HCV patients[81].

In summary, the current knowledge about the role of chemokines and their receptors during chronic hepatitis C strongly suggests that they are implicated in persistent liver inflammation. Moreover, HCV seems able to modulate the expression of some chemokine receptors through the induction of their ligands. This strategy could be used by HCV as a survival mechanism. First of all, it could impair specific T cell migration to the infected liver during the primary infection, weakening HCV clearing. Later, the mechanism could interfere in non-specific T cell migration to the liver during the chronic phase of infection to extend host survival. From a practical point of view, some chemokines and their receptors have been shown to be prognostic tools in predicting anti-HCV treatment responses, and after new larger studies to re-confirm these data, these predictors could be added to daily clinical practice. Finally, the blocking of chemokines and chemokine receptor engagement is a therapeutic strategy that should be explored in the near future for non-responders to current anti-HCV therapy.

Footnotes

Peer reviewers: Dr. Deborah L Diamond, Department of Microbiology, University of Washington, Box 358070, Seattle, WA 98195-8070, United States; Dr. Markus Reiser, Professor, Gastroenterology-Hepatology, Ruhr-University, Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany

S- Editor Li LF L- Editor O’Neill M E- Editor Zheng XM

References
1.  Lauer GM, Walker BD. Hepatitis C virus infection. N Engl J Med. 2001;345:41-52.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Afdhal NH. The natural history of hepatitis C. Semin Liver Dis. 2004;24 Suppl 2:3-8.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Manns MP, McHutchison JG, Gordon SC, Rustgi VK, Shiffman M, Reindollar R, Goodman ZD, Koury K, Ling M, Albrecht JK. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet. 2001;358:958-965.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Fried MW, Shiffman ML, Reddy KR, Smith C, Marinos G, Gonçales FL Jr, Häussinger D, Diago M, Carosi G, Dhumeaux D. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N Engl J Med. 2002;347:975-982.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Guidotti LG, Chisari FV. Immunobiology and pathogenesis of viral hepatitis. Annu Rev Pathol. 2006;1:23-61.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Lechner F, Wong DK, Dunbar PR, Chapman R, Chung RT, Dohrenwend P, Robbins G, Phillips R, Klenerman P, Walker BD. Analysis of successful immune responses in persons infected with hepatitis C virus. J Exp Med. 2000;191:1499-1512.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Thimme R, Oldach D, Chang KM, Steiger C, Ray SC, Chisari FV. Determinants of viral clearance and persistence during acute hepatitis C virus infection. J Exp Med. 2001;194:1395-1406.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Thimme R, Bukh J, Spangenberg HC, Wieland S, Pemberton J, Steiger C, Govindarajan S, Purcell RH, Chisari FV. Viral and immunological determinants of hepatitis C virus clearance, persistence, and disease. Proc Natl Acad Sci USA. 2002;99:15661-15668.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Bertoletti A, Maini MK. Protection or damage: a dual role for the virus-specific cytotoxic T lymphocyte response in hepatitis B and C infection? Curr Opin Immunol. 2000;12:403-408.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med. 2006;354:610-621.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Cerwenka A, Morgan TM, Harmsen AG, Dutton RW. Migration kinetics and final destination of type 1 and type 2 CD8 effector cells predict protection against pulmonary virus infection. J Exp Med. 1999;189:423-434.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Murai M, Yoneyama H, Harada A, Yi Z, Vestergaard C, Guo B, Suzuki K, Asakura H, Matsushima K. Active participation of CCR5(+)CD8(+) T lymphocytes in the pathogenesis of liver injury in graft-versus-host disease. J Clin Invest. 1999;104:49-57.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Maini MK, Boni C, Lee CK, Larrubia JR, Reignat S, Ogg GS, King AS, Herberg J, Gilson R, Alisa A. The role of virus-specific CD8(+) cells in liver damage and viral control during persistent hepatitis B virus infection. J Exp Med. 2000;191:1269-1280.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Price DA, Klenerman P, Booth BL, Phillips RE, Sewell AK. Cytotoxic T lymphocytes, chemokines and antiviral immunity. Immunol Today. 1999;20:212-216.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Luster AD. Chemokines--chemotactic cytokines that mediate inflammation. N Engl J Med. 1998;338:436-445.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol. 2004;22:891-928.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Gerard C, Rollins BJ. Chemokines and disease. Nat Immunol. 2001;2:108-115.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Bacon K, Baggiolini M, Broxmeyer H, Horuk R, Lindley I, Mantovani A, Maysushima K, Murphy P, Nomiyama H, Oppenheim J. Chemokine/chemokine receptor nomenclature. J Interferon Cytokine Res. 2002;22:1067-1068.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, Power CA. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol Rev. 2000;52:145-176.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Lodi PJ, Garrett DS, Kuszewski J, Tsang ML, Weatherbee JA, Leonard WJ, Gronenborn AM, Clore GM. High-resolution solution structure of the beta chemokine hMIP-1 beta by multidimensional NMR. Science. 1994;263:1762-1767.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Laudanna C, Campbell JJ, Butcher EC. Role of Rho in chemoattractant-activated leukocyte adhesion through integrins. Science. 1996;271:981-983.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Bonecchi R, Bianchi G, Bordignon PP, D'Ambrosio D, Lang R, Borsatti A, Sozzani S, Allavena P, Gray PA, Mantovani A. Differential expression of chemokine receptors and chemotactic responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med. 1998;187:129-134.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Qin S, Rottman JB, Myers P, Kassam N, Weinblatt M, Loetscher M, Koch AE, Moser B, Mackay CR. The chemokine receptors CXCR3 and CCR5 mark subsets of T cells associated with certain inflammatory reactions. J Clin Invest. 1998;101:746-754.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Mantovani A. The chemokine system: redundancy for robust outputs. Immunol Today. 1999;20:254-257.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Nansen A, Christensen JP, Andreasen SØ, Bartholdy C, Christensen JE, Thomsen AR. The role of CC chemokine receptor 5 in antiviral immunity. Blood. 2002;99:1237-1245.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Loetscher P, Uguccioni M, Bordoli L, Baggiolini M, Moser B, Chizzolini C, Dayer JM. CCR5 is characteristic of Th1 lymphocytes. Nature. 1998;391:344-345.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Napoli J, Bishop GA, McGuinness PH, Painter DM, McCaughan GW. Progressive liver injury in chronic hepatitis C infection correlates with increased intrahepatic expression of Th1-associated cytokines. Hepatology. 1996;24:759-765.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Bertoletti A, D'Elios MM, Boni C, De Carli M, Zignego AL, Durazzo M, Missale G, Penna A, Fiaccadori F, Del Prete G. Different cytokine profiles of intraphepatic T cells in chronic hepatitis B and hepatitis C virus infections. Gastroenterology. 1997;112:193-199.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Zeremski M, Petrovic LM, Talal AH. The role of chemokines as inflammatory mediators in chronic hepatitis C virus infection. J Viral Hepat. 2007;14:675-687.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell. 1994;76:301-314.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Butcher EC, Picker LJ. Lymphocyte homing and homeostasis. Science. 1996;272:60-66.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Patzwahl R, Meier V, Ramadori G, Mihm S. Enhanced expression of interferon-regulated genes in the liver of patients with chronic hepatitis C virus infection: detection by suppression-subtractive hybridization. J Virol. 2001;75:1332-1338.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Shields PL, Morland CM, Salmon M, Qin S, Hubscher SG, Adams DH. Chemokine and chemokine receptor interactions provide a mechanism for selective T cell recruitment to specific liver compartments within hepatitis C-infected liver. J Immunol. 1999;163:6236-6243.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Narumi S, Tominaga Y, Tamaru M, Shimai S, Okumura H, Nishioji K, Itoh Y, Okanoue T. Expression of IFN-inducible protein-10 in chronic hepatitis. J Immunol. 1997;158:5536-5544.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Larrubia JR, Calvino M, Benito S, Sanz-de-Villalobos E, Perna C, Pérez-Hornedo J, González-Mateos F, García-Garzón S, Bienvenido A, Parra T. The role of CCR5/CXCR3 expressing CD8+ cells in liver damage and viral control during persistent hepatitis C virus infection. J Hepatol. 2007;47:632-641.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Bièche I, Asselah T, Laurendeau I, Vidaud D, Degot C, Paradis V, Bedossa P, Valla DC, Marcellin P, Vidaud M. Molecular profiling of early stage liver fibrosis in patients with chronic hepatitis C virus infection. Virology. 2005;332:130-144.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Apolinario A, Majano PL, Alvarez-Pérez E, Saez A, Lozano C, Vargas J, García-Monzón C. Increased expression of T cell chemokines and their receptors in chronic hepatitis C: relationship with the histological activity of liver disease. Am J Gastroenterol. 2002;97:2861-2870.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Apolinario A, Diago M, Lo Iacono O, Lorente R, Pérez C, Majano PL, Clemente G, García-Monzón C. Increased circulating and intrahepatic T-cell-specific chemokines in chronic hepatitis C: relationship with the type of virological response to peginterferon plus ribavirin combination therapy. Aliment Pharmacol Ther. 2004;19:551-562.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Apolinario A, Majano PL, Lorente R, Núñez O, Clemente G, García-Monzón C. Gene expression profile of T-cell-specific chemokines in human hepatocyte-derived cells: evidence for a synergistic inducer effect of cytokines and hepatitis C virus proteins. J Viral Hepat. 2005;12:27-37.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Sillanpää M, Kaukinen P, Melén K, Julkunen I. Hepatitis C virus proteins interfere with the activation of chemokine gene promoters and downregulate chemokine gene expression. J Gen Virol. 2008;89:432-443.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Sprengers D, van der Molen RG, Kusters JG, Kwekkeboom J, van der Laan LJ, Niesters HG, Kuipers EJ, De Man RA, Schalm SW, Janssen HL. Flow cytometry of fine-needle-aspiration biopsies: a new method to monitor the intrahepatic immunological environment in chronic viral hepatitis. J Viral Hepat. 2005;12:507-512.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Leroy V, Vigan I, Mosnier JF, Dufeu-Duchesne T, Pernollet M, Zarski JP, Marche PN, Jouvin-Marche E. Phenotypic and functional characterization of intrahepatic T lymphocytes during chronic hepatitis C. Hepatology. 2003;38:829-841.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Harvey CE, Post JJ, Palladinetti P, Freeman AJ, Ffrench RA, Kumar RK, Marinos G, Lloyd AR. Expression of the chemokine IP-10 (CXCL10) by hepatocytes in chronic hepatitis C virus infection correlates with histological severity and lobular inflammation. J Leukoc Biol. 2003;74:360-369.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Helbig KJ, Ruszkiewicz A, Semendric L, Harley HA, McColl SR, Beard MR. Expression of the CXCR3 ligand I-TAC by hepatocytes in chronic hepatitis C and its correlation with hepatic inflammation. Hepatology. 2004;39:1220-1229.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Kusano F, Tanaka Y, Marumo F, Sato C. Expression of C-C chemokines is associated with portal and periportal inflammation in the liver of patients with chronic hepatitis C. Lab Invest. 2000;80:415-422.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115:209-218.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Zeremski M, Petrovic LM, Chiriboga L, Brown QB, Yee HT, Kinkhabwala M, Jacobson IM, Dimova R, Markatou M, Talal AH. Intrahepatic levels of CXCR3-associated chemokines correlate with liver inflammation and fibrosis in chronic hepatitis C. Hepatology. 2008;48:1440-1450.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Friedman SL. Liver fibrosis -- from bench to bedside. J Hepatol. 2003;38 Suppl 1:S38-S53.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Promrat K, McDermott DH, Gonzalez CM, Kleiner DE, Koziol DE, Lessie M, Merrell M, Soza A, Heller T, Ghany M. Associations of chemokine system polymorphisms with clinical outcomes and treatment responses of chronic hepatitis C. Gastroenterology. 2003;124:352-360.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Lloyd AR, Jagger E, Post JJ, Crooks LA, Rawlinson WD, Hahn YS, Ffrench RA. Host and viral factors in the immunopathogenesis of primary hepatitis C virus infection. Immunol Cell Biol. 2007;85:24-32.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Alter HJ. HCV natural history: the retrospective and prospective in perspective. J Hepatol. 2005;43:550-552.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Gruener NH, Lechner F, Jung MC, Diepolder H, Gerlach T, Lauer G, Walker B, Sullivan J, Phillips R, Pape GR. Sustained dysfunction of antiviral CD8+ T lymphocytes after infection with hepatitis C virus. J Virol. 2001;75:5550-5558.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Wiegand J, Cornberg M, Aslan N, Schlaphoff V, Sarrazin C, Kubitschke A, Buggisch P, Ciner A, Jaeckel E, Manns MP. Fate and function of hepatitis-C-virus-specific T-cells during peginterferon-alpha2b therapy for acute hepatitis C. Antivir Ther. 2007;12:303-316.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Lichterfeld M, Leifeld L, Nischalke HD, Rockstroh JK, Hess L, Sauerbruch T, Spengler U. Reduced CC chemokine receptor (CCR) 1 and CCR5 surface expression on peripheral blood T lymphocytes from patients with chronic hepatitis C infection. J Infect Dis. 2002;185:1803-1807.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Linnen J, Wages J Jr, Zhang-Keck ZY, Fry KE, Krawczynski KZ, Alter H, Koonin E, Gallagher M, Alter M, Hadziyannis S. Molecular cloning and disease association of hepatitis G virus: a transfusion-transmissible agent. Science. 1996;271:505-508.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Nattermann J, Nischalke HD, Kupfer B, Rockstroh J, Hess L, Sauerbruch T, Spengler U. Regulation of CC chemokine receptor 5 in hepatitis G virus infection. AIDS. 2003;17:1457-1462.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Xiang J, George SL, Wünschmann S, Chang Q, Klinzman D, Stapleton JT. Inhibition of HIV-1 replication by GB virus C infection through increases in RANTES, MIP-1alpha, MIP-1beta, and SDF-1. Lancet. 2004;363:2040-2046.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Williams CF, Klinzman D, Yamashita TE, Xiang J, Polgreen PM, Rinaldo C, Liu C, Phair J, Margolick JB, Zdunek D. Persistent GB virus C infection and survival in HIV-infected men. N Engl J Med. 2004;350:981-990.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Pileri P, Uematsu Y, Campagnoli S, Galli G, Falugi F, Petracca R, Weiner AJ, Houghton M, Rosa D, Grandi G. Binding of hepatitis C virus to CD81. Science. 1998;282:938-941.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Nattermann J, Nischalke HD, Feldmann G, Ahlenstiel G, Sauerbruch T, Spengler U. Binding of HCV E2 to CD81 induces RANTES secretion and internalization of CC chemokine receptor 5. J Viral Hepat. 2004;11:519-526.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Butera D, Marukian S, Iwamaye AE, Hembrador E, Chambers TJ, Di Bisceglie AM, Charles ED, Talal AH, Jacobson IM, Rice CM. Plasma chemokine levels correlate with the outcome of antiviral therapy in patients with hepatitis C. Blood. 2005;106:1175-1182.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Lagging M, Romero AI, Westin J, Norkrans G, Dhillon AP, Pawlotsky JM, Zeuzem S, von Wagner M, Negro F, Schalm SW. IP-10 predicts viral response and therapeutic outcome in difficult-to-treat patients with HCV genotype 1 infection. Hepatology. 2006;44:1617-1625.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Diago M, Castellano G, García-Samaniego J, Pérez C, Fernández I, Romero M, Iacono OL, García-Monzón C. Association of pretreatment serum interferon gamma inducible protein 10 levels with sustained virological response to peginterferon plus ribavirin therapy in genotype 1 infected patients with chronic hepatitis C. Gut. 2006;55:374-379.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Romero AI, Lagging M, Westin J, Dhillon AP, Dustin LB, Pawlotsky JM, Neumann AU, Ferrari C, Missale G, Haagmans BL. Interferon (IFN)-gamma-inducible protein-10: association with histological results, viral kinetics, and outcome during treatment with pegylated IFN-alpha 2a and ribavirin for chronic hepatitis C virus infection. J Infect Dis. 2006;194:895-903.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Zeremski M, Markatou M, Brown QB, Dorante G, Cunningham-Rundles S, Talal AH. Interferon gamma-inducible protein 10: a predictive marker of successful treatment response in hepatitis C virus/HIV-coinfected patients. J Acquir Immune Defic Syndr. 2007;45:262-268.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Yang YF, Tomura M, Iwasaki M, Ono S, Zou JP, Uno K, Shearer GM, Fujiwara H, Hamaoka T. IFN-alpha acts on T-cell receptor-triggered human peripheral leukocytes to up-regulate CCR5 expression on CD4+ and CD8+ T cells. J Clin Immunol. 2001;21:402-409.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Yamauchi K, Akbar SM, Horiike N, Michitaka K, Onji M. Increased serum levels of macrophage inflammatory protein-3alpha in chronic viral hepatitis: prognostic importance of macrophage inflammatory protein-3alpha during interferon therapy in chronic hepatitis C. J Viral Hepat. 2002;9:213-220.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Helbig KJ, George J, Beard MR. A novel I-TAC promoter polymorphic variant is functional in the presence of replicating HCV in vitro. J Clin Virol. 2005;32:137-143.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Woitas RP, Ahlenstiel G, Iwan A, Rockstroh JK, Brackmann HH, Kupfer B, Matz B, Offergeld R, Sauerbruch T, Spengler U. Frequency of the HIV-protective CC chemokine receptor 5-Delta32/Delta32 genotype is increased in hepatitis C. Gastroenterology. 2002;122:1721-1728.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Hellier S, Frodsham AJ, Hennig BJ, Klenerman P, Knapp S, Ramaley P, Satsangi J, Wright M, Zhang L, Thomas HC. Association of genetic variants of the chemokine receptor CCR5 and its ligands, RANTES and MCP-2, with outcome of HCV infection. Hepatology. 2003;38:1468-1476.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Wald O, Pappo O, Ari ZB, Azzaria E, Wiess ID, Gafnovitch I, Wald H, Spengler U, Galun E, Peled A. The CCR5Delta32 allele is associated with reduced liver inflammation in hepatitis C virus infection. Eur J Immunogenet. 2004;31:249-252.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Schols D. HIV co-receptors as targets for antiviral therapy. Curr Top Med Chem. 2004;4:883-893.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Huber M, Olson WC, Trkola A. Antibodies for HIV treatment and prevention: window of opportunity? Curr Top Microbiol Immunol. 2008;317:39-66.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Lederman MM, Penn-Nicholson A, Cho M, Mosier D. Biology of CCR5 and its role in HIV infection and treatment. JAMA. 2006;296:815-826.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Gong X, Feng H, Zhang S, Yu Y, Li J, Wang J, Guo B. Increased expression of CCR5 in experimental autoimmune myocarditis and reduced severity induced by anti-CCR5 monoclonal antibody. J Mol Cell Cardiol. 2007;42:781-791.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Suzuki K, Kawauchi Y, Palaniyandi SS, Veeraveedu PT, Fujii M, Yamagiwa S, Yoneyama H, Han GD, Kawachi H, Okada Y. Blockade of interferon-gamma-inducible protein-10 attenuates chronic experimental colitis by blocking cellular trafficking and protecting intestinal epithelial cells. Pathol Int. 2007;57:413-420.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Singh UP, Singh S, Taub DD, Lillard JW Jr. Inhibition of IFN-gamma-inducible protein-10 abrogates colitis in IL-10-/- mice. J Immunol. 2003;171:1401-1406.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Mohan K, Issekutz TB. Blockade of chemokine receptor CXCR3 inhibits T cell recruitment to inflamed joints and decreases the severity of adjuvant arthritis. J Immunol. 2007;179:8463-8469.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Singh UP, Venkataraman C, Singh R, Lillard JW Jr. CXCR3 axis: role in inflammatory bowel disease and its therapeutic implication. Endocr Metab Immune Disord Drug Targets. 2007;7:111-123.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Barth H, Robinet E, Liang TJ, Baumert TF. Mouse models for the study of HCV infection and virus-host interactions. J Hepatol. 2008;49:134-142.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Lalezari J, Yadavalli GK, Para M, Richmond G, Dejesus E, Brown SJ, Cai W, Chen C, Zhong J, Novello LA. Safety, pharmacokinetics, and antiviral activity of HGS004, a novel fully human IgG4 monoclonal antibody against CCR5, in HIV-1-infected patients. J Infect Dis. 2008;197:721-727.  [PubMed]  [DOI]  [Cited in This Article: ]