Hostname: page-component-7bb8b95d7b-495rp Total loading time: 0 Render date: 2024-09-27T06:38:16.037Z Has data issue: false hasContentIssue false

Effects of co-infection with Clonorchis sinensis on T cell exhaustion levels in patients with chronic hepatitis B

Published online by Cambridge University Press:  24 January 2024

Huimin Dong
Affiliation:
Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
Yuan Liao
Affiliation:
Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
Mei Shang
Affiliation:
Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
Yuechun Fu
Affiliation:
Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
Hongbin Zhang
Affiliation:
Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
Minqi Luo
Affiliation:
Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
Bo Hu*
Affiliation:
Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
*
Corresponding author: Bo Hu; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

To investigate the effects of co-infection with Clonorchis sinensis (C. sinensis) on T cell exhaustion levels in patients with chronic hepatitis B, we enrolled clinical cases in this study, including the patients with concomitant C. sinensis and HBV infection. In this study, we detected inhibitory receptors and cytokine expression in circulating CD4+ and CD8+ T cells by flow cytometry. PD-1 and TIM-3 expression levels were significantly higher on CD4+ T and CD8+ T cells from co-infected patients than on those from the HBV patients. In addition, CD4+ T cells and CD8+ T cells function were significantly inhibited by C. sinensis and HBV co-infection compared with HBV single infection, secreting lower levels of Interferon gamma (IFN-γ), Interleukin-2 (IL-2), and TNF-α. Our current results suggested that C. sinensis co-infection could exacerbate T cell exhaustion in patients with chronic hepatitis B. PD-1 and TIM-3 could be novel biomarkers for T cell exhaustion in patients with Clonorchis sinensis and chronic hepatitis B co-infection. Furthermore, it may be one possible reason for the weaker response to antiviral therapies and the chronicity of HBV infection in co-infected patients. We must realize the importance of C. sinensis treatment for HBV-infected patients. It might provide useful information for clinical doctors to choose the right treatment plans.

Type
Research Paper
Copyright
© The Author(s), 2024. Published by Cambridge University Press

Introduction

Clonorchiasis, resulted from Clonorchis sinensis (C. sinensis), is a food-borne parasitic disease. Over 35 million people are infected by C. sinensis in Asia, among which about 15 million infected people are prevalent in China (Botelho et al. Reference Botelho, Ferreira, Oliveira, Domingues, Machado and Da Costa2009; Deng et al. Reference Deng, Fang, Zhang, Mao, Pei and Liu2020; Na et al. Reference Na, Pak and Hong2020; Wang et al. Reference Wang, Young, Korhonen and Gasser2018). C. sinensis is a fish-borne trematode that locates in the bile duct of mammals, including humans (Young et al. Reference Young, Campbell, Hall, Jex, Cantacessi, Laha, Sohn, Sripa, Loukas, Brindley and Gasser2010). Freshwater fish act as the intermediate hosts in which the cercariae penetrate and transform into the metacercariae. Humans become infected by eating raw or undercooked freshwater fish (Liang et al. Reference Liang, Hu, Lv, Wu, Yu, Xu and Zheng2009).

Clinically, clonorchiasis patients exhibit different severity of the symptoms. Most of the infected people are not treated in time because of mild or unspecific symptoms, such as asthenia, nausea, diarrhea, jaundice, hepatomegaly, and liver tenderness. So, Clonorchiasis is a major but surprisingly neglected tropical disease in China. Only about 10% of the infected patients who have been treated in time show obvious acute clinical symptoms in the early infection, such as cholelithiasis, cholangitis, and cholecystitis. What is worse, chronic infection can develop into the cirrhosis or cholangiocarcinoma (Choi et al. Reference Choi, Han, Hong and Lee2004; Qian et al. Reference Qian, Chen, Liang, Yang and Zhou2012; Tang et al. Reference Tang, Huang and Yu2016).

Hepatitis B virus (HBV) infection is a major public health challenge that may cause severe complications, such as cirrhosis and hepatocellular carcinoma (HCC) (Trépo et al. Reference Trépo, Chan and Lok2014). Both C. sinensis infection and chronic hepatitis B virus infection can cause liver diseases. So, some studies have shown that the cases of the concurrent infection of HBV and C. sinensis often appear in areas where C. sinensis is prevalent, especially in the Guangdong province (Chen et al. Reference Chen, Xu, Zhou, Song, Wang and Zhu2012; Yang et al. Reference Yang, Han, Chen, Xu, Chen, Wang, Liu, Sun, Hou, Ning and Wang2014). Our previous study confirmed that co-infected patients presented weaker liver function and higher HBV Deoxyribo Nucleic Acid (DNA) copies, and the presence of C. sinensis may aggravate the disease state (Li et al. Reference Li, Dong, Huang, Chen, Kong, Sun, Yu and Xu2016a).

In chronic infections and cancer, T cells show poor effector function and persistent expression of multiple inhibitory receptors. This distinct state is often associated with T cell dysfunction, which is described as ‘exhaustion’ (Bertoletti A and Naoumov NV Reference Bertoletti and Naoumov2003; Chisari et al. Reference Chisari, Isogawa and Wieland2010). T cell exhaustion was first described during chronic lymphocytic choriomeningitis virus (LCMV) infection in mice more than a decade ago as dysfunction of antigen-specific T cells (Gallimore et al. Reference Gallimore, Glithero, Godkin, Tissot, Plückthun, Elliott, Hengartner and Zinkernagel1998; Zajac et al. Reference Zajac, Blattman, Murali-Krishna, Sourdive, Suresh, Altman and Ahmed1998). Since then, it has been confirmed in plenty of animal models and humans with chronic viral infections and cancer (Virgin et al. Reference Virgin, Wherry and Ahmed2009).

T cell exhaustion is characterised by the expression of multiple cell surface inhibitory receptors, defective production of some cytokines, and appearance of immunoregulatory cells, such as programmed cell death-1 (PD-1), lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin mucin-3 (Tim-3), cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), Interleukin-2 (IL-2), Interferon gamma (IFN-γ), and transforming growth factor-α (TNF-α) (Evans et al. Reference Evans, Riva, Cooksley, Phillips, Puranik, Nathwani, Brett, Chokshi and Naoumov2008; Hartzell et al. Reference Hartzell, Bin, Cantarelli, Haverly, Manrique, Angeletti, Manna, Murphy, Zhang, Levitsky, Gallon, Yu and Cravedi2020; Liu et al. Reference Liu, Gao, Liang and Ma2016; Wongjitrat et al. Reference Wongjitrat, Sukwit, Chuenchitra, Seangjaruk, Rojanasang, Romputtan and Srisurapanon2013; Ye et al. Reference Ye, Li, Dong, Wang, Tian, Lin, Liu, Kong and Chen2017).

Many studies have illustrated that T cell exhaustion has also been observed in humans during chronic infection diseases, such as HBV, hepatitis C virus (HCV), LCMV, human immunodeficiency virus (HIV), and cancers (Boni et al. Reference Boni, Laccabue, Lampertico, Giuberti, Viganò, Schivazappa, Alfieri, Pesci, Gaeta, Brancaccio, Colombo, Missale and Ferrari2012; Fisicaro et al. Reference Fisicaro, Barili, Rossi, Montali, Vecchi, Acerbi, Laccabue, Zecca, Penna, Missale, Ferrari and Boni2020; Saeidi et al. Reference Saeidi, Zandi, Cheok, Saeidi, Wong, Lee, Cheong, Yong, Larsson and Shankar2018; Ye et al. Reference Ye, Liu, Li, Kong, Tian and Chen2015). It has been linked to these chronic infectious diseases. Our study puts attention on T cell exhaustion in HBV infection. HBV cannot be cleaned up, and continuous replication then develops into liver cirrhosis and liver cancer. The main reason for the chronicity of HBV infection is virus-specific T cell exhaustion (Bertoletti and Ferrari Reference Bertoletti and Ferrari2012; Ferrari Reference Ferrari2020; Meng et al., Reference Meng, Chen and Lu2019). Reversal of these exhausted T cells will pave the way for the development of more effective immunotherapeutic strategies for the treatment of chronic HBV infection.

The aim of our study was to evaluate the effect of co-infection with C. sinensis on T cell exhaustion levels in patients with chronic hepatitis B. Our results showed that C. sinensis co-infection could exacerbate T cell exhaustion in patients with chronic hepatitis B. Furthermore, it may be one possible reason for the weaker response to antiviral therapies in co-infected patients. We must realize the importance of C. sinensis treatment for HBV-infected patients. It might provide useful information for clinical doctors to choose the right treatment plans.

Materials and methods

Subjects

All patients were hospitalised or followed up at the Third Affiliated Hospital of Sun Yat-sen University. According to the infection of HBV and C. sinensis, the enrolled subjects were divided into four groups: patients who were mono-infected with HBV surface-antigen (HBsAg)-positive and HBV DNA >20 IU/mL, patients who were mono-infected with C. sinensis eggs-positive and patients who were co-infected with HBsAg-positive, HBV DNA >20 IU/mL and C. sinensis eggs-positive, healthy individuals with matched age and sex as normal controls.

The inclusion criteria for normal controls were negative for both HBsAg and C. sinensis eggs. In addition, mono-infected patients with HBV and co-infected patients with HBV and C. sinensis were prescribed antiviral drugs, entecavir (ETV, 0.5 mg once daily) only. Neither mono-infected with C. sinensis nor co-infected patients with HBV and C. sinensis were treated with anthelmintic treatment.

The exclusion criterion for all the enrolled subjects with the following other causes of chronic liver damage were excluded from the study: hepatitis A, C, D, and E, HIV, Schistosoma japonicum, Schistosoma mansoni or other parasites, or alcohol. And Patients with autoimmune diseases, diabetes, hematological system diseases, serious heart diseases, or pregnant women were also excluded from the study. Twenty-five healthy individuals with matched age and sex were also enrolled as normal controls.

We summarised the clinical characteristics obtained for the enrolled subjects in Table 1. The study protocol was approved by the Clinical Research Ethics Committee of the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China, and informed consent forms were signed by all participants according to the Declaration of Helsinki.

Table 1. Clinical characteristics of the study groups

* Data are expressed as the Mean ± SD or median and ranges. AST: aspartate aminotransferase; ALT: alanine aminotransferase; COI: cut-off index; DNA: Deoxyribo Nucleic Acid; HBV: hepatitis B virus; HC: healthy control; TB: total bilirubin; SD: Standard Deviation; a: statistically significantly different versus healthy control group; b: statistically significantly different versus HBV mono-infected group; c: statistically significantly different versus C. sinensis mono-infected group.

Virological and biochemical assessments

Biochemical assessments were measured using automated biochemical techniques (Hitachi 7600, Tokyo, Japan). The serum HBsAg level was detected using the electrochemiluminescence immunoassay kit for the cobas e801 system (Roche Diagnostics, Mannheim, Germany), with a positive result recorded as cut-off index (COI) > 1.00. The serum levels of HBV DNA were determined by real-time PCR with a lower detection limit of 20 IU/ml (Roche Diagnostics, Mannheim, Germany)

Eggs of C. sinensis per gram of feces count

Eggs of C. sinensis per gram of feces were determined by a sodium hydroxide digestion method in mono-infected and co-infected with C. sinensis patients. The specific process of sodium hydroxide digestion is as follows: 1 g feces is put into a centrifugal sedimentation tube containing 5 ml of 10% sodium hydroxide solution, then we stir it fully, and digest it for 24 hr. We suck 0.075 ml for smear and count the eggs in the whole piece under the microscope, and then multiply it by 100.

Peripheral blood mononuclear cell isolation

Peripheral blood mononuclear cells (PBMCs) were from fresh heparinised blood (5 ml) collected from each group of patients and isolated by Ficoll-Hypaque density gradient centrifugation (Haoyang Biological Manufacture, Tianjin, China). Cell viability was analysed by trypan blue staining and automated counting of live and dead cells. We ensured that the viability of PBMCs was more than 90% after isolation. PBMCs were cultured at 1×106 cells/ml in Roswell Park Memorial Institute (RPMI) 1640 (Invitrogen Gibco, New York, USA) supplemented with 10% fetal bovine serum (FBS; Invitrogen Gibco).

Determination of PD-1, TIM-3, LAG-3, and CTLA-4 expression by flow cytometry

The PBMCs were washed twice with a Phosphate Buffered Saline (PBS) buffer and incubated with Anti-CD3-PE-Cy7 (eBioscience, California, USA), anti-CD4-APC (eBioscience, California, USA), anti-CD8-FITC (eBioscience, California, USA), anti-PD-1-PerCP-Cy5.5 (eBioscience, California, USA), anti-Tim-3-PE (eBioscience, California, USA), anti-LAG-3-A1eXa Fluor⑩647 (eBioscience, California, USA), and anti-CTLA-4-BV421 (eBioscience, California, USA) for surface staining at room temperature for 30 min in the dark. IgG isotype control antibodies were used as the negative controls. Stained cells were analysed with a BD FACS Aria II analyser (BD Biosciences, New Jersey, USA), and data were analysed with FlowJo version 10 for Windows (Tree Star Inc., California, USA).

Determination of intracellular cytokine release by flow cytometry

The PBMCs were stimulated with a cell stimulation cocktail (eBioscience, California, USA), which is a cocktail of phorbol 12-myristate 13-acetate (PMA) and ionomycin for 4 hr, followed by the addition of the protein transport inhibitor cocktail (eBioscience, California, USA), a cocktail of Brefeldin A and monensin, at 2 hr before detection. After 6 hr of incubation, anti-CD3-PE-Cy7 (eBioscience, California, USA), anti-CD4-APC (eBioscience, California, USA), and anti-CD8-FITC (eBioscience, California, USA) were used for cell staining at room temperature for 30 min in the dark. Intracellular staining for cytokines was performed with a Fixation/Permeabilization Kit (BD Biosciences, New Jersey, USA) and after permeabilisation and fixation, the cells were washed twice with a PBS buffer and incubated with anti-IFN-γ-Alexa Fluor 647 (Biolegend, California, USA), anti-IL-2-BV42 (Biolegend, California, USA), and anti-TNF-α-PE (Biolegend, California, USA) at room temperature for 30 min in the dark. IgG isotype control antibodies were used as the negative controls. Staining cells were analysed with a BD FACS Aria II analyser (BD Biosciences, New Jersey, USA), and data were analysed with FlowJo version 10 for Windows (Tree Star Inc., California, USA).

Statistical analysis

All data of continuous variables were presented as the mean values ± standard error or median and ranges. Differences in continuous variables were performed by one-way Analysis of Variance (ANOVA) for comparison with more than two groups. The Wilcoxon rank sum test was used for non-parametric data. The data were analysed using GraphPad Prism 7.0. P values < 0.05 were considered statistically significant.

Results

Demographic and clinical characteristics of the study subjects

The enrolled subjects in this study were classified into four groups: co-infected group (n = 20), HBV group (n = 24), C. sinensis group (n = 23), and HC group (n = 25). Age, gender, liver function, HBsAg, and serum HBV DNA concentrations are demonstrated in Table 1. The co-infected group showed higher levels of aspartate aminotransferase (ALT), alanine aminotransaminase (AST), and total bilirubin (TB) than the C. sinensis group and the HC group. Especially, the levels of ALT, AST, and TB in the co-infected group were significantly higher than that in the HBV group (P < 0.05, respectively). However, no difference in the HBV DNA log copies and HBsAg was found in the co-infected group and in the HBV group (P > 0.05). In brief, these data indicate that C. sinensis may weaken liver function and aggravate the disease state in patients with chronic hepatitis B. Eventually, C. sinensis and HBV co-infection may lead to the chronicity of HBV infection.

Expression of inhibitory receptors on the surface of peripheral blood CD4+ T and CD8+ T cells from the co-infected group, HBV group and C. sinensis group, and the HC group

The frequencies of CD4+ T and CD8+ T cells with surface expression of the inhibitory receptors PD-1, TIM-3, LAG-3, and CTLA-4 were evaluated in the co-infected group, HBV group, C. sinensis group, and HC group using flow cytometry. Compared with the HC group, the percentage of PD-1+CD4+ T cells, PD-1+CD8+ T cells, TIM-3+CD4+ T cells, TIM-3+CD8+ T cells, LAG-3+CD8+ T cells, CTLA-4+CD4+ T cells, and CTLA-4+CD8+ T cells were all significantly increased in both the HBV group and C. sinensis group (all P < 0.005, Figure 1), while no significant difference was found in the LAG-3 on the CD4+ T cells (Figure 1b). Significantly higher frequencies of PD-1+CD4+, PD-1+CD8+, TIM-3+CD4+, and TIM-3+CD8+ cells were observed in the co-infected group than in the HBV group (P = 0.000, P = 0.017, P = 0.001, and P = 0.002, respectively). No difference was found in the LAG-3 and CTLA-4 expression levels in both the CD4+ T cells and CD8+ T cells obtained from the co-infected group than those in the CD4+ T cells and CD8+ T cells obtained from the HBV group (Figure 1). This result demonstrated that high PD-1 and TIM-3 expression in the co-infected group could exacerbate T cell exhaustion in patients with chronic hepatitis B.

Figure 1. Gating strategy and expression profiles of PD-1, TIM-3, LAG-3, and CTLA-4 in CD4+ T and CD8+ T cells in the co-infected group, HBV group, C. sinensis group, and HC group. (A) The gating strategies and representative results of PD-1, TIM-3, LAG-3, and CTLA-4 expression in CD4+ T and CD8+ T cells. (B) The percentages of PD-1, TIM-3, LAG-3, and CTLA-4 in CD4+ T cells expression from co-infected group (n = 20), HBV group (n = 24), C. sinensis group (n = 23), and HC group (n = 25). (C) The percentages of PD-1, TIM-3, LAG-3, and CTLA-4 in CD8+ T cells expression from the four groups. Data show the means ± SD. Asterisks indicate statistically significant differences between two groups, as measured by one-way ANOVA (*P < 0.05, **P < 0.01, ***P < 0.001).

Difference in cytokines production between the CD4+ T and CD8+ T cells in the co-infected group, HBV group and C. sinensis group, and the HC group

The percentages of CD4+ T and CD8+ T cells with cytokine expression, such as IL-2, IFN-γ, and TNF-α, were assessed in the four groups by flow cytometry. Compared with the HC group, the percentage of IFN-γ, IL-2, and TNF-α in both the CD4+ T cells and CD8+ T cells were all significantly decreased in both the HBV group and co-infected group (all P < 0.01, Figure 2), and lower IL-2 expression levels were detected in both the CD4+ T cells and CD8+ T cells obtained from the co-infected group than those in the CD4+ T cells and CD8+ T cells obtained from the HBV group (P = 0.006, P = 0.033, Figure 2b, 2c). The co-infected group showed lower levels of IFN-γ and TNF-α in the CD8+ T cells than the HBV group (P = 0.047, P = 0.046, Figure 2c), while no significant difference was found in the IFN-γ and TNF-α in the CD4+ T cells. (Figure 2b). These findings suggested that the CD4+ T cells and CD8+ T cells function was inhibited by C. sinensis and HBV co-infection, secreting low IFN-γ, IL-2, and TNF-α upon stimulation with a cell stimulation cocktail and the protein transport inhibitor cocktail separately.

Figure 2. Gating strategy and expression profiles of IL-2, IFN-γ, and TNF-α in CD4+ T and CD8+ T cells in the co-infected group, HBV group, C. sinensis group, and HC group. (A) The gating strategies and representative results of IL-2, IFN-γ, and TNF-α expression in CD4+ T and CD8+ T cells. (B) The percentages of IL-2, IFN-γ, and TNF-α expression in CD4+ T cells expression from the co-infected group (n = 20), HBV group (n = 24), C. sinensis group (n = 23), and HC group (n = 25). (C) The percentages of IL-2, IFN-γ, and TNF-α expression in CD8+ T cell expression from the four groups. Data show the means ± SD. Asterisks indicate statistically significant differences between two groups, as measured by one-way ANOVA (*P < 0.05, **P < 0.01, ***P < 0.001).

Discussion

T cell exhaustion is considered as a general characteristic of chronic viral infections and cancer, such as HBV (Wherry Reference Wherry2011). A large amount of studies have confirmed that T cell exhaustion has also been observed in humans during chronic infection diseases (Nebbia et al. Reference Nebbia, Peppa, Schurich, Khanna, Singh, Cheng, Rosenberg, Dusheiko, Gilson, ChinAleong, Kennedy and Maini2012; Wang et al. Reference Wang, He, Shen, Lu and Sun2019a; Wang et al. Reference Wang, Zheng, Shi and Xu2019b). As a consequence, the immunoregulation of T cell exhaustion in chronic HBV infection will provide different therapeutic targets and molecular controls to treat chronic HBV infection.

C. sinensis infection can evolve into a sustained chronic condition. Because trematode locates in the bile duct of the host, it can lead to several chronic diseases, such as periductal liver fibrosis, cholangitis, and even hepatic cirrhosis or cholangiocarcinoma. C. sinensis infection was also closely related to T cell exhaustion. Our previous study investigated the relationship between HBV infection and C. sinensis infection, and the presence of C. sinensis may aggravate the disease state. In this study, we provided further evidence that C. sinensis co-infection could exacerbate T cell exhaustion in patients with chronic hepatitis B.

During HBV infection and clearance, it is well known that CD8+ T cells play a key role. However, CD4+ T cells are the key factor regulating on the cellular cytotoxic T lymphocyte (CTL) response to HBV because they are needed for the development of the optimal effector CTL and for the generation and maintenance of functional memory CTL (Fisicaro et al. Reference Fisicaro, Boni, Barili, Laccabue and Ferrari2018; Wherry Reference Wherry2011). According to previous research, the lack of CD4+ T cells was the main cause of CD8+ T cell exhaustion (Trautmann et al. Reference Trautmann, Kozik, Carambia, Richter, Lischke, Schwinge, Mittrücker, Lohse, Oxenius, Wiegard and Herkel2014). Like CD8+ T cells, specific CD4+ T cells also lose effector function during chronic viral infection. Therefore, longer persistent infection or loss of help from CD4+ T cells leads to more severe T cell exhaustion (Wherry and Ahmed, Reference Wherry and Ahmed2004). Although T cell exhaustion of CD4+ T cells is important, little research has been conducted on CD4+ T cell exhaustion in chronic HBV-infected patients (Antoine et al. Reference Antoine, Varner, Carville, Connole, Marchant and Kaur2014). This may be the reason that our study focused on both CD4+ T cells and CD8+ T cells.

Actually, T cells exhibited progressive and gradual exhaustion during persistent chronic infection (Jackson et al. Reference Jackson, Berrien-Elliott, Meyer, Wherry and Teague2013), and it is characterised by an upregulation in the expression of inhibitory molecules, such as PD-1, TIM-3, LAG-3, and CTLA-4. All the results from our study were in accordance with the previous reports. Our data showed that the cell surface inhibitory receptors, such as PD-1, TIM-3, LAG-3, and CTLA-4, levels were expressed significantly higher by CD8+ T cells in the HBV group than in the HC group. CD4+ T cells expressed higher PD-1, TIM-3, and CTLA-4 levels, not including LAG-3, in the HBV group than in the HC group. In addition, we also analysed the PD-1, TIM-3, LAG-3, and CTLA-4 expression levels on both CD4+ T cells and CD8+ T cells among the C. sinensis mono-infected patients. These inhibitory receptors were expressed higher in the C. sinensis group relative to the expression levels in the HC group. Our results provide strong evidence that the two infectious factors of hepatitis B and C. sinensis would eventually lead to T cell exhaustion.

The numbers and types of inhibitory receptors are closely related to the degree of T cell exhaustion (Nguyen and Ohashi, Reference Nguyen and Ohashi2015; Yoshio et al., Reference Yoshio, Sugiyama, Shoji, Mano, Mita, Okamoto, Matsuura, Okuno, Takikawa, Mizokami and Kanto2016). Many studies have demonstrated the relationship between the extent of T cell exhaustion and the severity of infection (Streeck et al. Reference Streeck, Brumme, Anastario, Cohen, Jolin, Meier, Brumme, Rosenberg, Alter, Allen, Walker and Altfeld2008; Wherry et al. Reference Wherry, Blattman, Murali-Krishna, van der Most and Ahmed2003a, Reference Wherry, Blattman and Ahmed2005). In this study, we focused on the exhausted T cells, especially those in patients with co-infection C. sinensis and HBV. C. sinensis co-infection could exacerbate T cell exhaustion in patients with chronic hepatitis B. Our data showed that PD-1 and TIM-3 levels were expressed significantly higher on CD4+ T cells and CD8+ T cells in the co-infected group than those in the HBV group. Furthermore, the recovery of T cell function is increased considerably by simultaneous block of the PD-1 pathway, LAG-3 pathway, CTLA-4 pathway, and TIM-3 pathway (Jin et al. Reference Jin, Anderson, Tan, West, Ha, Araki, Freeman, Kuchroo and Ahmed2010; Kaufmann et al. Reference Kaufmann, Kavanagh, Pereyra, Zaunders, Mackey, Miura, Palmer, Brockman, Rathod, Piechocka-Trocha, Baker, Zhu, Le Gall, Waring, Ahern, Moss, Kelleher, Coffin, Freeman, Rosenberg and Walker2007; Nakamoto et al. Reference Nakamoto, Cho, Shaked, Olthoff, Valiga, Kaminski, Gostick, Price, Freeman, Wherry and Chang2009). Meanwhile, the findings from our study showed only PD-1 and TIM-3 levels were significantly increased on the surface of exhausted T cells in the co-infected group than those in the HBV group.

Among all the inhibitory receptors expressed on the surface of exhausted T cells, PD-1 was considered as one of the best-known inhibitory receptors (Bengsch et al. Reference Bengsch, Martin and Thimme2014). The axis of PD-1 and its ligand seems to be a major inhibitory receptor pathway in T cell exhaustion. Many studies confirmed that in vivo blockade of PD-1 leads to a substantial improvement in virus-specific CD8+ T cell responses and also enhancement of B cell responses. Therefore, blocking the PD-1 pathway could be a major immunotherapeutic strategy for achieving immunological control of diseases in humans (Brahmer et al. Reference Brahmer, Drake, Wollner, Powderly, Picus, Sharfman, Stankevich, Pons, Salay, McMiller, Gilson, Wang, Selby, Taube, Anders, Chen, Korman, Pardoll, Lowy and Topalian2010; Velu et al. Reference Velu, Titanji, Zhu, Husain, Pladevega, Lai, Vanderford, Chennareddi, Silvestri, Freeman, Ahmed and Amara2009). Recently, it was reported that Tim-3 had a non-redundant role in chronic infected patients similar to PD-1 because Tim-3 and PD-1 may be expressed on overlapping subsets of T cells, and Tim-3 was treated as another inhibitory marker of exhausted T cells during chronic infection. The high expressions of Tim-3 levels were correlated with the state of CD8+ T cell exhaustion, induction of T cell dysfunction, and suppression of natural killer cells (Golden-Mason et al., Reference Golden-Mason, Palmer, Kassam, Townshend-Bulson, Livingston, McMahon, Castelblanco, Kuchroo, Gretch and Rosen2009; Jones et al., Reference Jones, Ndhlovu, Barbour, Sheth, Jha, Long, Wong, Satkunarajah, Schweneker, Chapman, Gyenes, Vali, Hyrcza, Yue, Kovacs, Sassi, Loutfy, Halpenny, Persad, Spotts, Hecht, Chun, McCune, Kaul, Rini, Nixon and Ostrowski2008). In addition, our data showed that no difference was found in the LAG-3 and CTLA-4 expression levels. Further investigations are required to address this point. It may be due to the small number of these cases. T cell exhaustion leads to a weakened or suppressed host immune response, which is a status of gradual T cell dysfunction that arises during chronic infections (Schietinger and Greenberg Reference Schietinger and Greenberg2014). So, immunoregulation is centrally involved.

The main performances are not only the previous inhibitory receptors but also the inhibitory cytokines (Wherry and Kurachi, Reference Wherry and Kurachi2015). T cells mainly secrete cytokines, such as IL-2, IFN-γ, and TNF-α, to eradicate viruses and parasites causing intracellular infections and play an important role in cellular immunity (Shaw et al. Reference Shaw, Merien, Braakhuis and Dulson2018). We sought to understand the T cell exhaustion, as measured by changes in the IFN-γ, IL-2, and TNF-α secretion. The data demonstrated that lower IFN-γ, IL-2, and TNF-α expression levels were detected in both the CD4+ T cells and CD8+ T cells from both the CHB group and C. sinensis group than in the T cells obtained from the HC group. The results suggest that the two infectious factors of hepatitis B and C. sinensis would eventually lead to T cell exhaustion. At the same time, IFN-γ, IL-2, and TNF-α expression levels were detected as significantly lower in the CD8+ T cells from the co-infected group than those in the CD8+ T cells obtained from the HBV group. But in CD4+ T cells, only IL-2 expression levels were detected as significantly lower from the co-infected group than those from the HBV group. Obviously, C. sinensis has more effect on cytokines levels in the CD8+ T cells from the co-infected group. Significantly, during T cell exhaustion, the downregulation of secretion of IFN-γ, IL-2, and TNF-α occurs in a hierarchical manner. It was a process with decreasing IL-2 secretion first, followed by the loss of IFN-γ and TNF-α production (Crawford and Wherry Reference Crawford and Wherry2009; Wherry et al. Reference Wherry, Ha, Kaech, Haining, Sarkar, Kalia, Subramaniam, Blattman, Barber and Ahmed2007). Many researchers put their attention on IL-2. Previous studies have highlighted that IL-2 promotes the formation of effector CD8+ T cells, and supplementing IL-2 can reverse T cell exhaustion and recover T cell proliferation (Bachmann et al. Reference Bachmann, Wolint, Walton, Schwarz and Oxenius2007; Pipkin et al. Reference Pipkin, Sacks, Cruz-Guilloty, Lichtenheld, Bevan and Rao2010; Schwartz Reference Schwartz2003). According to these studies and our study, it is reasonable to believe that IL-2 is the most important cytokine to prevent T cell exhaustion.

Patients with chronic HBV infections are usually characterised by a population of exhausted T cells, which have weak virus-specific T cell responses, impeding the clearance of the virus and recovery from hepatitis. Continuously high viral load and high antigen levels may contribute to liver injury and inflammation and the exhaustion of HBV-specific T cells. So, there is a close relationship between liver damage and T cell exhaustion, as our experimental data also confirm this point.

In recent years, there has been more and more research that shows the mechanisms explaining the exhausted T cells in patients who develop chronic HBV infection (Wherry Reference Wherry2011; Bertoletti and Gehring Reference Bertoletti and Gehring2006). These mechanisms for the development of T cell exhaustion were not only observed in chronic HBV infection but also have been confirmed in chronic parasitic infection. It has been proposed that some mechanisms may contribute to the dysfunction of T cells, such as continuously high viral load and high antigen levels, suppressive cytokines including IFN-γ and TNF-α, and dendritic cells, which were able to lead to a progressive exhausted T cell function (Goncalves et al. 2010; Karp et al. Reference Karp, El-Safi, Wynn, Satti, Kordofani, Hashim, Hag-Ali, Neva, Nutman and Sacks1993).

Overall, the present research on the mechanism of T cell exhaustion is based almost entirely on viral models. Although some mechanisms have been proven in some parasitic diseases, the significance of these molecules during T cell exhaustion needs to be studied more thoroughly in chronic parasitic models. Our study may show that the interaction between C. sinensis and hepatitis B virus may exacerbate T cell exhaustion in patients with chronic hepatitis B. Considering that chronic C. sinensis and hepatitis B virus co-infection can have a potentially additive effect on T cell exhaustion, we should pay more attention to the T cell exhaustion caused by C. sinensis infection in the co-infection patients. Overall, our research provides direction for understanding the mechanisms of T cell exhaustion and for developing effective immunotherapy strategies for these diseases.

Our study may suggest that C. sinensis co-infection could exacerbate T cell exhaustion in patients with chronic hepatitis B. This is consistent with the previous notion that in co-infected patients, the efficacy of antiviral treatment was better in patients who were prescribed with entecavir and praziquantel than entecavir alone. This result further indicated that the efficacy of HBV antiviral treatment was related to the removal of worms in co-infected patients. One possible reason for the weaker response to antiviral therapies in co-infected patients could therefore be exacerbated T cell exhaustion in this group.

Conclusions

In summary, this study is the first to provide strong evidence that C. sinensis co-infection could exacerbate T cell exhaustion in patients with chronic hepatitis B. C. sinensis and HBV co-infection may lead to the chronicity of HBV infection, and C. sinensis may play a role in the unresponsiveness to antiviral therapy in co-infected patients. We must realise the importance of C. sinensis treatment for HBV-infected patients. PD-1 and TIM-3 might be used as novel biomarkers for T cell exhaustion in patients with Clonorchis sinensis and chronic hepatitis B co-infection.

Acknowledgments

We are thankful to all volunteers who participated in this study.

Financial support

This work was supported by grants from the National Natural Science Foundation of China [grant number 81902082].

Competing interest

None.

Ethical standard

The study ethical standards were approved by the Clinical Research Ethics Committee of the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China (permission number 0204501).

Footnotes

Huimin Dong and Yuan Liao contributed equally to this work.

References

Antoine, P, Varner, V, Carville, A, Connole, M, Marchant, A and Kaur, A (2014) Postnatal acquisition of primary rhesus cytomegalovirus infection is associated with prolonged virus shedding and impaired CD4+ T lymphocyte function. Journal of Infectious Diseases 210(7), 10901099. https://doi.org/10.1093/infdis/jiu215CrossRefGoogle ScholarPubMed
Bachmann, MF, Wolint, P, Walton, S, Schwarz, K and Oxenius, A (2007) Differential role of IL-2R signaling for CD8+ T cell responses in acute and chronic viral infections. European Journal of Immunology 37(6), 15021512. https://doi.org/10.1002/eji.200637023CrossRefGoogle ScholarPubMed
Bengsch, B, Martin, B and Thimme, R (2014) Restoration of HBV-specific CD8+ T cell function by PD-1 blockade in inactive carrier patients is linked to T cell differentiation. Journal of Hepatology 61(6), 12121219. https://doi.org/10.1016/j.jhep.2014.07.005CrossRefGoogle ScholarPubMed
Bertoletti, A and Naoumov, NV (2003) Translation of immunological knowledge into better treatments of chronic hepatitis B. Journal of Hepatology 39(1), 115124. https://doi.org/10.1016/S0168-8278(03)00126-0CrossRefGoogle ScholarPubMed
Bertoletti, A and Gehring, AJ (2006) The immune response during hepatitis B virus infection. Journal of General Virology 87(6), 14391449. https://doi.org/10.1099/vir.0.81920-0CrossRefGoogle ScholarPubMed
Bertoletti, A and Ferrari, C (2012) Innate and adaptive immune responses in chronic hepatitis B virus infections: Towards restoration of immune control of viral infection. Postgraduate Medical Journal 89(1051), 294304. https://doi.org/10.1136/gutjnl-2011-301073CrossRefGoogle Scholar
Boni, C, Laccabue, D, Lampertico, P, Giuberti, T, Viganò, M, Schivazappa, S, Alfieri, A, Pesci, M, Gaeta, GB, Brancaccio, G, Colombo, M, Missale, G and Ferrari, C (2012) Restored function of HBV-specific T cells after long-term effective therapy with nucleos(t)ide analogues. Gastroenterology 143(4), 963973. https://doi.org/10.1053/j.gastro.2012.07.014CrossRefGoogle ScholarPubMed
Botelho, M, Ferreira, AC, Oliveira, MJ, Domingues, A, Machado, JC and Da Costa, JMC (2009) Schistosoma haematobium total antigen induces increased proliferation, migration and invasion, and decreases apoptosis of normal epithelial cells. International Journal for Parasitology 39(10), 10831091. https://doi.org/10.1016/j.ijpara.2009.02.016CrossRefGoogle ScholarPubMed
Brahmer, JR, Drake, CG, Wollner, I, Powderly, JD, Picus, J, Sharfman, WH, Stankevich, E, Pons, A, Salay, TM, McMiller, TL, Gilson, MM, Wang, C, Selby, M, Taube, JM, Anders, R, Chen, L, Korman, AJ, Pardoll, DM, Lowy, I and Topalian, SL (2010) Phase I study of single-agent anti–programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity, pharmacodynamics, and immunologic correlates. Journal of Clinical Oncology 28(19), 31673175. https://doi.org/10.1200/JCO.2009.26.7609CrossRefGoogle ScholarPubMed
Chen, J, Xu, M, Zhou, D, Song, H, Wang, C and Zhu, X (2012) Canine and feline parasitic zoonoses in China. Parasites & Vectors 5(1), 152. https://doi.org/10.1186/1756-3305-5-152CrossRefGoogle ScholarPubMed
Chisari, FV, Isogawa, M and Wieland, SF (2010) Pathogenesis of hepatitis B virus infection. Patholologie Biologie (Paris) 58(4), 258266. https://doi.org/10.1016/j.patbio.2009.11.001CrossRefGoogle ScholarPubMed
Choi, BI, Han, JK, Hong, ST and Lee, KH (2004) Clonorchiasis and cholangiocarcinoma: Etiologic relationship and imaging diagnosis. Clinical Microbiology Reviews 17(3), 540552. https://doi.org/10.1128/CMR.17.3.540-552.2004CrossRefGoogle ScholarPubMed
Crawford, A and Wherry, EJ (2009) The diversity of costimulatory and inhibitory receptor pathways and the regulation of antiviral T cell responses. Current Opinion in Immunology 21(2), 179186. https://doi.org/10.1016/j.coi.2009.01.010CrossRefGoogle ScholarPubMed
Deng, ZH, Fang, YY, Zhang, QM, Mao, Q, Pei, FQ and Liu, MR (2020) The control of clonorchiasis in Guangdong Province, southern China. Acta Tropica 202, 105246. https://doi.org/10.1016/j.actatropica.2019.105246CrossRefGoogle ScholarPubMed
Evans, A, Riva, A, Cooksley, H, Phillips, S, Puranik, S, Nathwani, A, Brett, S, Chokshi, S and Naoumov, NV (2008) Programmed death 1 expression during antiviral treatment of chronic hepatitis B: Impact of hepatitis B e-antigen seroconversion. Hepatology 48(3), 759769. https://doi.org/10.1002/hep.22419CrossRefGoogle ScholarPubMed
Ferrari, C (2020) HBV and the immune response. Liver International 35(Suppl 1), 121128. https://doi.org/10.1111/liv.12749CrossRefGoogle Scholar
Fisicaro, P, Boni, C, Barili, V, Laccabue, D and Ferrari, C (2018) Strategies to overcome HBV-specific T cell exhaustion: Checkpoint inhibitors and metabolic re-programming. Current Opinion in Virology 30, 18. https://doi.org/10.1016/j.coviro.2018.01.003CrossRefGoogle ScholarPubMed
Fisicaro, P, Barili, V, Rossi, M, Montali, I, Vecchi, A, Acerbi, G, Laccabue, D, Zecca, A, Penna, A, Missale, G, Ferrari, C and Boni, C (2020) Pathogenetic mechanisms of T cell dysfunction in chronic HBV infection and related therapeutic approaches. Frontiers in Immunology 11, 849. https://doi.org/10.3389/fimmu.2020.00849CrossRefGoogle ScholarPubMed
Gallimore, A, Glithero, A, Godkin, A, Tissot, AC, Plückthun, A, Elliott, T, Hengartner, H and Zinkernagel, R (1998) Induction and exhaustion of lymphocytic choriomeningitis virus-specific cytotoxic T lymphocytes visualized using soluble tetrameric major histocompatibility complex class I-peptide complexes. Journal of Experimental Medicine 187(9), 13831393. https://doi.org/10.1084/jem.187.9.1383CrossRefGoogle ScholarPubMed
Golden-Mason, L, Palmer, BE, Kassam, N, Townshend-Bulson, L, Livingston, S, McMahon, BJ, Castelblanco, N, Kuchroo, V, Gretch, DR and Rosen, HR (2009) Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. Journal of Virology 83(18), 91229130. https://doi.org/10.1128/JVI.00639-09.CrossRefGoogle Scholar
Gonçalves, RM, Salmazi, KC, Santos, BAN, Bastos, MS, Rocha, SC, Boscardin, SB, Silber, AM, Kallás, EG, Ferreira, MU and Scopel, KKG (2010) CD4+ CD25+ Foxp3+ Regulatory T cells, dendritic cells, and circulating cytokines in uncomplicated malaria: Do different parasite species elicit similar host responses? Infection and Immunity 78(11), 47634772. https://doi.org/10.1128/IAI.00578-10CrossRefGoogle ScholarPubMed
Hartzell, S, Bin, S, Cantarelli, C, Haverly, M, Manrique, J, Angeletti, A, Manna, G, Murphy, B, Zhang, W, Levitsky, J, Gallon, L, Yu, SM and Cravedi, P (2020) Kidney failure associates with T cell exhaustion and imbalanced follicular helper T cells. Frontiers in Immunology 11, 583702. https://doi.org/10.3389/fimmu.2020.583702CrossRefGoogle Scholar
Jackson, SR, Berrien-Elliott, MM, Meyer, JM, Wherry, EJ and Teague, RM (2013) CD8+ T cell exhaustion during persistent viral infection is regulated independently of the virus-specific T cell receptor. Immunological Investigations 42(3), 204220. https://doi.org/10.3109/08820139.2012.751397CrossRefGoogle ScholarPubMed
Jin, HT, Anderson, AC, Tan, WG, West, EE, Ha, SJ, Araki, K, Freeman, GJ, Kuchroo, VK and Ahmed, R (2010) Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proceedings of the National Academy of Sciences USA 107(33), 1473314738. https://doi.org/10.1073/pnas.1009731107CrossRefGoogle ScholarPubMed
Jones, RB, Ndhlovu, LC, Barbour, JD, Sheth, PM, Jha, AR, Long, BR, Wong, JC, Satkunarajah, M, Schweneker, M, Chapman, JM, Gyenes, G, Vali, B, Hyrcza, MD, Yue, FY, Kovacs, C, Sassi, A, Loutfy, M, Halpenny, R, Persad, D, Spotts, G, Hecht, FM, Chun, T, McCune, JM, Kaul, R, Rini, JM, Nixon, DF and Ostrowski, MA (2008) Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. The Journal of Experimental Medicine 205(12), 27632779. https://doi.org/10.1084/jem.20081398CrossRefGoogle ScholarPubMed
Karp, CL, El-Safi, SH, Wynn, TA, Satti, MM, Kordofani, AM, Hashim, FA, Hag-Ali, M, Neva, FA, Nutman, TB and Sacks, DL (1993) In vivo cytokine profiles in patients with kala-azar. Marked elevation of both interleukin-10 and interferon-gamma. The Journal of Clinical Investigation 91(4), 16441648. https://doi.org/10.1172/JCI116372CrossRefGoogle ScholarPubMed
Kaufmann, DE, Kavanagh, DG, Pereyra, F, Zaunders, JJ, Mackey, EW, Miura, T, Palmer, S, Brockman, M, Rathod, A, Piechocka-Trocha, A, Baker, B, Zhu, B, Le Gall, S, Waring, MT, Ahern, R, Moss, K, Kelleher, AD, Coffin, JM, Freeman, GJ, Rosenberg, ES and Walker, BD (2007) Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat Immunol 8(11), 12461254. https://doi.org/10.1038/ni1515CrossRefGoogle ScholarPubMed
Li, W, Dong, H, Huang, Y, Chen, T, Kong, X, Sun, H, Yu, X and Xu, J (2016a) Clonorchis sinensis co-infection could affect the disease state and treatment response of HBV patients. PLOS Neglected Tropical Diseases 10(6), e4806. https://doi.org/10.1371/journal.pntd.0004806CrossRefGoogle ScholarPubMed
Liang, C, Hu, XC, Lv, ZY, Wu, ZD, Yu, XB, Xu, J and Zheng, HQ (2009) [Experimental establishment of life cycle of Clonorchis sinensis]. Zhongguo Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi 27(2), 148150. http://doi.org/CNKI:SUN:ZJSB.0.2009-02-019Google ScholarPubMed
Liu, Y, Gao, LF, Liang, XH and Ma, CH (2016) Role of Tim-3 in hepatitis B virus infection: An overview. World Journal of Gastroenterology 22(7), 22942303. https://doi.org/10.3748/wjg.v22.i7.2294CrossRefGoogle ScholarPubMed
Lykken, JM, DiLillo, DJ, Weimer, ET, Roser-Page, S, Heise, MT, Grayson, JM, Weitzmann, MN and Tedder, TF (2014) Acute and chronic B cell depletion disrupts CD4+ and CD8+ T cell homeostasis and expansion during acute viral infection in mice. Journal of Immunology (Baltimore, Md.: 1950) 193(2), 746756. https://doi.org/10.4049/jimmunol.1302848CrossRefGoogle ScholarPubMed
Meng, Z, Chen, Y and Lu, M (2019) Advances in targeting the innate and adaptive immune systems to cure chronic hepatitis B virus infection. Frontiers in Immunology 10, 3127. https://doi.org/10.3389/fimmu.2019.03127CrossRefGoogle ScholarPubMed
Na, BK, Pak, JH and Hong, SJ (2020) Clonorchis sinensis and clonorchiasis. Acta Tropica 203, 105309. https://doi.org/10.1016/j.actatropica.2019.105309CrossRefGoogle ScholarPubMed
Nakamoto, N, Cho, H, Shaked, A, Olthoff, K, Valiga, ME, Kaminski, M, Gostick, E, Price, DA, Freeman, GJ, Wherry, EJ and Chang, KM (2009) Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathogens 5(2), e1000313. https://doi.org/10.1371/journal.ppat.1000313CrossRefGoogle ScholarPubMed
Nebbia, G, Peppa, D, Schurich, A, Khanna, P, Singh, HD, Cheng, Y, Rosenberg, W, Dusheiko, G, Gilson, R, ChinAleong, J, Kennedy, P and Maini, MK (2012) Upregulation of the Tim-3/galectin-9 pathway of T cell exhaustion in chronic hepatitis B virus infection. PloS One 7(10), e47648. https://doi.org/10.1371/journal.pone.0047648CrossRefGoogle ScholarPubMed
Nguyen, LT and Ohashi, PS (2015) Clinical blockade of PD1 and LAG3 — potential mechanisms of action. Nature Reviews Immunology 15(1), 4556. https://doi.org/10.1038/nri3790CrossRefGoogle ScholarPubMed
Pipkin, ME, Sacks, JA, Cruz-Guilloty, F, Lichtenheld, MG, Bevan, MJ and Rao, A (2010) Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity 32(1), 7990. https://doi.org/10.1016/j.immuni.2009.11.012CrossRefGoogle ScholarPubMed
Qian, MB, Chen, YD, Liang, S, Yang, GJ and Zhou, XN (2012) The global epidemiology of clonorchiasis and its relation with cholangiocarcinoma. Infectious Diseases of Poverty 1(1), 4. https://doi.org/10.1186/2049-9957-1-4CrossRefGoogle ScholarPubMed
Saeidi, A, Zandi, K, Cheok, YY, Saeidi, H, Wong, WF, Lee, C, Cheong, HC, Yong, YK, Larsson, M and Shankar, EM (2018) T-cell exhaustion in chronic infections: reversing the state of exhaustion and reinvigorating optimal protective immune responses. Frontiers in Immunology 9, 2569. https://doi.org/10.3389/fimmu.2018.02569CrossRefGoogle ScholarPubMed
Schietinger, A and Greenberg, PD (2014) Tolerance and exhaustion: Defining mechanisms of T cell dysfunction. Trends in Immunology 35(2), 5160. https://doi.org/10.1016/j.it.2013.10.001CrossRefGoogle ScholarPubMed
Shaw, DM, Merien, F, Braakhuis, A and Dulson, D (2018) T-cells and their cytokine production: The anti-inflammatory and immunosuppressive effects of strenuous exercise. Cytokine 104, 136142. https://doi.org/10.1016/j.cyto.2017.10.001CrossRefGoogle ScholarPubMed
Streeck, H, Brumme, ZL, Anastario, M, Cohen, KW, Jolin, JS, Meier, A, Brumme, CJ, Rosenberg, ES, Alter, G, Allen, TM, Walker, BD and Altfeld, M (2008) Antigen load and viral sequence diversification determine the functional profile of HIV-1-specific CD8+ T cells. PLoS Medicine 5(5), e100. https://doi.org/10.1371/journal.pmed.0050100CrossRefGoogle ScholarPubMed
Tang, ZL, Huang, Y and Yu, XB (2016) Current status and perspectives of Clonorchis sinensis and clonorchiasis: Epidemiology, pathogenesis, omics, prevention and control. Infect Dis Poverty 5(1), 71. https://doi.org/10.1186/s40249-016-0166-1CrossRefGoogle ScholarPubMed
Trautmann, T, Kozik, JH, Carambia, A, Richter, K, Lischke, T, Schwinge, D, Mittrücker, HW, Lohse, AW, Oxenius, A, Wiegard, C and Herkel, J (2014) CD4+ T-cell help is required for effective CD8+ T cell-mediated resolution of acute viral hepatitis in mice. PLoS One 9(1), e86348. https://doi.org/10.1371/journal.pone.0086348CrossRefGoogle ScholarPubMed
Trépo, C, Chan, HL and Lok, A (2014) Hepatitis B virus infection. The Lancet 384(9959), 20532063. https://doi.org/10.1016/S0140-6736(14)60220-8CrossRefGoogle ScholarPubMed
Velu, V, Titanji, K, Zhu, B, Husain, S, Pladevega, A, Lai, L, Vanderford, TH, Chennareddi, L, Silvestri, G, Freeman, GJ, Ahmed, R and Amara, RR (2009) Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 458(7235), 206210. https://doi.org/10.1038/nature07662CrossRefGoogle ScholarPubMed
Virgin, HW, Wherry, EJ and Ahmed, R (2009) Redefining chronic viral infection. Cell 138(1), 3050. https://doi.org/10.1016/j.cell.2009.06.036CrossRefGoogle ScholarPubMed
Wang, D, Young, ND, Korhonen, PK and Gasser, RB (2018) Clonorchis sinensis and Clonorchiasis: The relevance of exploring genetic variation. Adv Parasitol 100, 155208. https://doi.org/10.1016/bs.apar.2018.03.006CrossRefGoogle ScholarPubMed
Wang, X, He, Q, Shen, H, Lu, X and Sun, B (2019a) Genetic and phenotypic difference in CD8(+) T cell exhaustion between chronic hepatitis B infection and hepatocellular carcinoma. Journal of Medical Genetics 56(1), 1821. https://doi.org/10.1136/jmedgenet-2018-105267CrossRefGoogle ScholarPubMed
Wang, Y, Zheng, D, Shi, M and Xu, X (2019b) T cell dysfunction in chronic hepatitis B infection and liver cancer: Evidence from transcriptome analysis. Journal of Medical Genetics 56(1), 22. https://doi.org/10.1136/jmedgenet-2018-105570CrossRefGoogle ScholarPubMed
Wherry, EJ (2011) T cell exhaustion. Nature Immunology 12(6), 492499. https://doi.org/10.1038/ni.2035CrossRefGoogle ScholarPubMed
Wherry, EJ, Blattman, JN, Murali-Krishna, K, van der Most, R and Ahmed, R (2003a) Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. Journal of Virology 77(8), 49114927. https://doi.org/10.1128/jvi.77.8.4911-4927.2003CrossRefGoogle ScholarPubMed
Wherry, EJ, Blattman, JN and Ahmed, R (2005) Low CD8 T-cell proliferative potential and high viral load limit the effectiveness of therapeutic vaccination. Journal of Virology 79(14), 89608968. https://doi.org/10.1128/JVI.79.14.8960-8968.2005CrossRefGoogle ScholarPubMed
Wherry, EJ, Ha, S, Kaech, SM, Haining, WN, Sarkar, S, Kalia, V, Subramaniam, S, Blattman, JN, Barber, DL and Ahmed, R (2007) Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27(4), 670684. https://doi.org/10.1016/j.immuni.2007.09.006CrossRefGoogle ScholarPubMed
Wherry, EJ and Ahmed, R (2004) Memory CD8 T-cell differentiation during viral infection. Journal of Virology 78(11), 55355545. https://doi.org/10.1128/JVI.78.11.5535-5545.2004.CrossRefGoogle ScholarPubMed
Wherry, EJ and Kurachi, M (2015) Molecular and cellular insights into T cell exhaustion. Nature Reviews Immunology 15(8), 486499. https://doi.org/10.1038/nri3862CrossRefGoogle ScholarPubMed
Wongjitrat, C, Sukwit, S, Chuenchitra, T, Seangjaruk, P, Rojanasang, P, Romputtan, P and Srisurapanon, S (2013) CTLA-4 and its ligands on the surface of T- and B-lymphocyte subsets in chronic hepatitis B virus infection. Journal of the Medical Association of Thailand 96(Suppl 1), S54S59. https://europepmc.org/article/MED/23724456Google ScholarPubMed
Yang, G, Han, M, Chen, F, Xu, Y, Chen, E, Wang, X, Liu, Y, Sun, J, Hou, J, Ning, Q and Wang, Z (2014) Hepatitis B virus genotype B and mutations in basal core promoter and pre-core/core genes associated with acute-on-chronic liver failure: A multicenter cross-sectional study in China. Hepatology International 8(4), 508516. https://doi.org/10.1007/s12072-014-9554-4CrossRefGoogle ScholarPubMed
Ye, B, Liu, X, Li, X, Kong, H, Tian, L and Chen, Y (2015) T-cell exhaustion in chronic hepatitis B infection: Current knowledge and clinical significance. Cell Death & Disease 6(3), e1694. https://doi.org/10.1038/cddis.2015.42CrossRefGoogle ScholarPubMed
Ye, B, Li, X, Dong, Y, Wang, Y, Tian, L, Lin, S, Liu, X, Kong, H and Chen, Y (2017) Increasing LAG-3 expression suppresses T-cell function in chronic hepatitis B: A balance between immunity strength and liver injury extent. Medicine (Baltimore) 96(1), e5275. https://doi.org/10.1097/MD.0000000000005275CrossRefGoogle ScholarPubMed
Yoshio, S, Sugiyama, M, Shoji, H, Mano, Y, Mita, E, Okamoto, T, Matsuura, Y, Okuno, A, Takikawa, O, Mizokami, M and Kanto, T (2016) Indoleamine-2,3-dioxygenase as an effector and an indicator of protective immune responses in patients with acute hepatitis B. Hepatology 63(1), 8394. https://doi.org/10.1002/hep.28282CrossRefGoogle Scholar
Young, ND, Campbell, BE, Hall, RS, Jex, AR, Cantacessi, C, Laha, T, Sohn, WM, Sripa, B, Loukas, A, Brindley, PJ and Gasser, RB (2010) Unlocking the transcriptomes of two carcinogenic parasites, Clonorchis sinensis and Opisthorchis viverrini. PLoS Neglected Tropical Diseases 4(6), e719. https://doi.org/10.1371/journal.pntd.0000719CrossRefGoogle ScholarPubMed
Zajac, AJ, Blattman, JN, Murali-Krishna, K, Sourdive, DJ, Suresh, M, Altman, JD and Ahmed, R (1998) Viral immune evasion due to persistence of activated T cells without effector function. Journal of Experimental Medicine 188(12), 22052213. https://doi.org/10.1084/JEM.188.12.2205CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Clinical characteristics of the study groups

Figure 1

Figure 1. Gating strategy and expression profiles of PD-1, TIM-3, LAG-3, and CTLA-4 in CD4+ T and CD8+ T cells in the co-infected group, HBV group, C. sinensis group, and HC group. (A) The gating strategies and representative results of PD-1, TIM-3, LAG-3, and CTLA-4 expression in CD4+ T and CD8+ T cells. (B) The percentages of PD-1, TIM-3, LAG-3, and CTLA-4 in CD4+ T cells expression from co-infected group (n = 20), HBV group (n = 24), C. sinensis group (n = 23), and HC group (n = 25). (C) The percentages of PD-1, TIM-3, LAG-3, and CTLA-4 in CD8+ T cells expression from the four groups. Data show the means ± SD. Asterisks indicate statistically significant differences between two groups, as measured by one-way ANOVA (*P < 0.05, **P < 0.01, ***P < 0.001).

Figure 2

Figure 2. Gating strategy and expression profiles of IL-2, IFN-γ, and TNF-α in CD4+ T and CD8+ T cells in the co-infected group, HBV group, C. sinensis group, and HC group. (A) The gating strategies and representative results of IL-2, IFN-γ, and TNF-α expression in CD4+ T and CD8+ T cells. (B) The percentages of IL-2, IFN-γ, and TNF-α expression in CD4+ T cells expression from the co-infected group (n = 20), HBV group (n = 24), C. sinensis group (n = 23), and HC group (n = 25). (C) The percentages of IL-2, IFN-γ, and TNF-α expression in CD8+ T cell expression from the four groups. Data show the means ± SD. Asterisks indicate statistically significant differences between two groups, as measured by one-way ANOVA (*P < 0.05, **P < 0.01, ***P < 0.001).