Original Article Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Hepatol. Mar 27, 2011; 3(3): 72-78
Published online Mar 27, 2011. doi: 10.4254/wjh.v3.i3.72
Polymorphisms in programmed death-1 gene are not associated with chronic HBV infection in Chinese patients
Feng Lv, Yu-Feng Gao, Zhen-Huan Zhang, Ming-Fang Cui, Shu-Ling Xia, Xu Li, Hua-Fa Yin, Department of Infectious Diseases, the First Affiliated Hospital, Anhui Medical University, Hefei 230022, Anhui Province, China
Tian-Chen Zhang, Fa-Ming Pan, Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei 230032, Anhui Province, China
Author contributions: Lv F and Gao YF contributed equally to this work; Zhang ZH provided the vital reagents and analytical tools; Lv F and Gao YF wrote the manuscript; Lv F, Cui MF and Xia SL performed the majority of experiments; Zhang TC and Pan FM took charge of collecting all human materials; Yin HF designed the study; and Li X provided the financial support for this work.
Supported by the Grants from the National Natural Science Foundation of China, No.30700698 and 30771907 and the Foundation of Pre-973 Program Projects, No. 2009CB526411
Correspondence to: Hua-Fa Yin, Professor, Department of Infectious Diseases, the First Affiliated Hospital, Anhui Medical University, 218 Jixi Road, Hefei 230022, Anhui Province, China. yhf163.good@163.com
Telephone: +86-551-2922912 Fax: +86-551-2922912
Received: November 26, 2010
Revised: December 23, 2010
Accepted: December 30, 2010
Published online: March 27, 2011

Abstract

AIM: To investigate the association between the programmed death-1(PD-1) polymorphisms and genetic susceptibility of chronic hepatitis B virus (HBV) infection in Chinese patients.

METHODS: Two single nucleotide polymorphisms (SNPs), PD-1.1 G > A and PD-1.2 G > A, were genotyped in 539 patients with chronic HBV infection and 353 other family members (HbsAg-) from 256 nuclear families using polymerase chain reactiorestriction fragment length polymorphisms assay. The associations between PD-1 polymorphisms and genetic susceptibility of chronic HBV infection were analyzed usng the family-based association analysis method.

RESULTS: No association or linkage was detected among 539 patients. Univariate (single-marker) family-based association tests demonstrated that PD-1 genotypes, alleles and transmitted haplotypes are not associated with chronic HBV infection (all with P value more than 0.05). Transmission/disequilibrium test and sibship disequilibrium test analysis showed no excess of the alleles from heterozygous parents to affected offspring (P = 0.688880, P = 1.000000 respectively).

CONCLUSION: The data demonstrated that PD-1.1 and PD-1.2 polymorphisms are not associated with chronic HBV infection in Chinese patients.

Key Words: Programmed death-1, Hepatitis B virus, Single nucleotide polymorphism, Genetic association study, Family-based association test



INTRODUCTION

Chronic hepatitis B virus (HBV) infection has been considered a multi-factorial and polygenic disorder with viral, environmental, genetic and immune components[1-3]. However, segregation analysis and twin studies strongly support the role of host genetic components in determining the chronicity of HBV infection[4-6]. Genetic association analyses have implicated that some human leukocyte antigen and non-human leukocyte antigen loci, including interferon-γ (IFN-γ)[7], tumor necrosis factor-α (TNF-α)[8], cytotoxic T lymphocyte antigen 4 (CTLA-4)[9], estrogen receptor-α (ESR1)[10], type 1 interferon receptor 1(IFNR-1)[11] and so on, are associated with chronic HBV infection or HBV clearance. Susceptibility to infectious diseases is considered to be determined at different functional levels such as cytokine production, antigen presentation and receptor recognition. Therefore, studies on the chronicity of HBV infection association with certain genes involved in the immune response may highlight the genetic background of HBV infection.

Programmed cell death 1 (PD-1) is an inhibitory immunoreceptor expressed on T-cells, B-cells and myeloid cells. It belongs to the immunoglobulin super family B7/CD28 and is mainly involved in T cells and B cells activation[12,13]. In patients with HBV infection, PD-1 is significantly up-regulated on the virus specific T cells and leads to inhibition of T cell receptor-mediated proliferation and cytokine production[14,15], thereby playing a crucial role in the down-regulation of immune responses. The human gene encoding PD-1, i.e. PDCD1, is located on chromosome 2q37.3[16]. Until now, more than 30 single nucleotide polymorphisms (SNPs) have been identified within PD-1 gene (found in the NCBI-Entrez SNP database). Many reports have highlighted that some regulatory polymorphisms in PD-1 gene might affect the expression and transcription of the gene[17,18] such as in the promoter or intron; they have been studied as a part of attempts to identify the pathogenesis of several immune relevant diseases including systemic lupus erythematosus (SLE)[17,19], rheumatoid arthritis (RA)[20] and type1 diabetes (T1D)[21]. Further work has shown that other SNPs in the PD-1 gene were studied in multiple sclerosis (MS)[22], ankylosing spondylitis (AS)[23], Graves’ disease[24] and so on. However, no such studies have, until now, been performed within chronic HBV infection with a sufficiently large sample population. Hence, considering the important role of PD-1 in HBV-specific T cells response and the possible effects of expression or functional alteration of PD-1 due to gene polymorphisms on the immune response, our present study was firstly designed to investigate the association of PD-1 SNPs including PD-1.1 (position -531 in the promoter) and PD-1.2 (position 6438 in the intron 2) with genetic susceptibility of chronic HBV infection. Therefore,we performed a family-based association study in Chinese nuclear families to ascertain whether there is a susceptible gene locus for chronic HBV infection.

MATERIALS AND METHODS
Subjects

A family-based association study of chronic HBV infection was conducted in Anhui province, China between 2008 and 2009. All probands were diagnosed with chronic HBV infection and fulfilled the diagnostic criteria of the Proposal of Prevention and Treatment of Viral Hepatitis, Xi’an, 2000, issued by the Chinese Society of Infectious Diseases and Parasitology and the Chinese Society of Hepatology of the Chinese Medical Association[25]; they were positive for HBsAg and anti-HBc but negative for antibodies (Abs) to HCV, HDV, HIV-1 and -2, and excluded other symptoms of chronic liver damage. In a total of 892 subjects [539 patients (aged 36.5±14.6 years) and 353 other family members (aged 44.1±15.5 years)] from 256 qualified nuclear families [114 (44.53%) families had both parents, 94 (36.72%) families had one single parent and 48 (18.75%) families had no available parents] were finally recruited. The family criteria used for subject selection were as follows: (1) proband’s parents, (2) at least two siblings, and (3) at least one parent or one additional sibling. All subjects gave their written informed consent and blood and this study obtained ethical committee approval from the Department of Infectious Diseases, Anhui Medical University. Peripheral whole blood samples were collected in 5-ml vacationer tubes containing EDTA and detected serum HBsAg by enzyme-linked immunosorbent assay (ELIAS) kits (Kehua Technologies Co., Ltd. Shanghai, China).

Genomic DNA extraction

Genomic DNA was extracted from the peripheral blood leucocytes pellet using a DNA extraction kit (Yuan Ping-hao Biotechnology Co., Ltd. Tianjin, China). The DNA samples were stored at -80°C with a concentration of 100 ng/μL.

Genotyping by polymerase chain reaction and restriction fragment length polymorphisms analysis

Two SNPs (PD-1.1 G > A and PD-1.2 G > A) were selected as targets and were genotyped using the TaqMan system (Applied Biosystems). The PD-1.1 G > A primers were forward, 5’-GATCTGGAACTGTGGCCATGGT-3’ and reverse, 5’-CCCCCTCTGGGCTCAGGTT-3’, and those for PD-1.2 G > A were 5’-CGGTCCTGGGGTGGGTGTCC-3’ and 5’-GCTGGGGTGGGCTGTGGGCA-3’ respectively. They were amplified in a 25 μL reaction mixture which consisted of 3 μL DNA (100 ng/μL), 1 μL deoxynucleoside triphosphates (dNTP, 10 mmol/μL), 2.5 μL 10 × buffer, 0.5 μL of Taq polymerase (5 U/μL, TaKaRa Biotechnology, Co., Ltd. Dalian, China), 1 μL each primer (10 μmol/μL, Invitrogen Biotechnology Co., Ltd. Shanghai, China) and H2O 16 μL. The amplification conditions for determining PD-1.1G > A polymorphisms were initial denaturation at 95°C for 5 min and 30 cycles of denaturation at 95°C for 15 s, annealing at 67°C for 15 s, extension at 72°C for 15 s and then a final extension phase at 72°C for 10 min. The polymerase chain reaction (PCR) product 265 bp was digested with MspI (TaKaRa Biotechnology, Co., Ltd. Dalian, China) according to the manufacturer’s instructions [total reaction volume 20 μL, including PCR product 10 μL, 10× T buffer 2 μL, 0.1% BSA 2 μL, restriction enzyme (10 U/μL) 1μL, H2O 5 μL] and separated by electrophoresis on 3% agarose gels. The AA genotype was cleaved and appeared as 180 and 85 bp fragments and GG genotype appeared as 125, 85 and 55 bp fragments, whereas the AG genotype appeared as 180, 125, 85 and 55 bp fragments (Figure 1). To determine the polymorphisms of PD-1.2G > A, PCR was carried out under the following conditions: initial denaturation at 94°C for 5 min and 30 cycles of denaturation at 94°C for 40 s, annealing at 70°C for 15 s and extension at 72°C for 15 s. A final extension phase was performed at 72°C for 7 min. Then the PCR product was digested with MspI (TaKaRa Biotechnology Co., Ltd. Dalian, China) and separated by electrophoresis on 3% agarose gel. The AA genotype lacking the MspI site migrated as a 263 bp fragment whereas the GG genotype was cleaved and appeared as 150 and 113 bp. The AG genotype was digested into three fragments 263, 150 and 113 bp (Figure 1). To improve the genotyping quality and validate our results, a random selection of 1% of all samples were analyzed by direct sequencing (Invitrogen Biotechnology Co., Ltd, Shanghai, China) and the results found no discrepancies.

Figure 1
Figure 1 Gel electrophoresis patterns of PD-1. 1 and PD-1.2. A: The amplified fragments of PD-1.1 were digested with MspI: the polymerase chain reaction (PCR) product size was 265 base pairs (bp) which was digested to 180 and 85 bp, the genotype was identified as AA; if not, it was digested to 180, 125, 85 and 55 bp and was identified as AG; and if only digested to 125, 85 and 55 bp, its genotype was identified as GG. B: The amplified fragments of PD-1.2 were also digested with MspI: the PCR product size was 263 bp which was digested to 150 and 113 bp; if the product was digested, the genotype was identified as GG; if not, it was identified as AA, the genotype AG appeared as 263, 150 and 113 bp.
Statistical analysis

In order to avoid the bias of population admixture arising from population-based association study, an extension of the transmission/ disequilibrium test and unified family-based association test (FBAT)[26] were performed under dominant, recessive and additive genetic models. The number of informative families was dependent on the genetic models. The statistical power of FBAT depended on the number of informative families. The single-marker FBAT analysis was used to estimate the single loci frequencies. Each test counted how often a specific locus was present in informative families with HBV infection. Positive Z statistic of single locus FBAT indicated a specific single locus was more frequently transmitted to patients with HBV infection in informative families than expected under the null hypothesis of no linkage and no association. To test transmission disequilibrium in families, we applied the conventional TDT statistic (disequilibrium of transmission of alleles from heterozygous parents to affected children) and the SDT (considering patient and other normal siblings). Statistic is calculated as (b - c)2/(b + c), where b and c are, respectively, the number of transmissions and non-transmissions of the alleles. Haplotype FBAT software[27] was also used to estimate haplotype frequencies. For families with ambiguous phase, expectation-maximization algorithm[28] was used to estimate genotype frequencies under Hardy-Weinberg equilibrium.

RESULTS

A total of 539 patients with chronic HBV infection from 256 nuclear families that contained complete genotype were employed in the data analysis. At the same time, 353 family members of these patients were genotyped. Stratified by PD-1.1 G > A and PD-1.2 G > A genotypes among 539 patients, the frequencies of PD-1.1 G > A, AA, AG and GG genotypes were 16.6%, 63.7% and 19.7% respectively and the A and G allele frequencies were 48.4% and 51.6% respectively. PD-1.2 G > A, AA, AG and GG genotype frequencies were 22.8%, 59.8% and 17.4% respectively and the A and G allele frequencies were 52.7% and 47.3% respectively (Table 1). The genotype distributions of PD-1.1 and PD-1.2 polymorphisms were in Hardy-Weinberg equilibrium (PD-1.1, χ2 = 3.032, P = 0.082; PD-1.2, χ2 = 0.25, P = 0.616).

Table 1 The associations between PD-1 genotype and chronic HBV infection by family-based association test.
MarkerGenotypeFrequencyFamilyaSE(S)Var(S)ZbP
PD-1.1A/A0.166463029.31112.7180.1930.847
A/G0.637724749.79420.122-0.6230.533
G/G0.197423027.89511.6300.6170.537
PD-1.2A/A0.228533834.22814.2061.0010.317
A/G0.598785054.04421.696-0.8680.385
G/G0.174412726.72811.5930.0800.936

Univariate (single-marker) FBAT demonstrated that there were no associations between genotypes and chronic HBV infection (P > 0.05). In PD-1.1 G > A and PD-1.2 G > A, single loci analysis by FBAT showed that the alleles of A and G were also not associated with chronic HBV infection in the additive model [(PD-1.1, Z = -0.265, P = 0.790993; Z = 0.265, P = 0.790993 respectively); (PD-1.2, Z = 0.640, Z = -0.640, P = 0.522127 respectively)], the dominant model [(PD-1.1, Z = -0.617, P = 0.537018; Z = -0.193, P = 0.846892 respectively); (PD-1.2, Z = -0.080, P = 0.936343; Z = -1.001, P = 0.316939 respectively)] and the recessive model [(PD-1.1, Z = 0.193, P = 0.846892; Z = 0.617, P = 0.537018 respectively); (PD-1.2, Z = 1.001, P = 0.316939; Z = 0.080, P = 0.936343 respectively)]. The results of single-marker FBAT analyses were summarized in Table 2. TDT and SDT analysis revealed no increased transmission for the major alleles from heterozygous parents to affected offspring (P = 0.688880, P = 1.000000 respectively) (Table 3).

Table 2 The associations between PD-1 gene polymorphism and chronic HBV infection by family-based association test (single-marker FBAT analysis) in additive or dominant or recessive model.
MarkerAlleleAdditive modelDominant modelRecessive model
Familiesa
Zb
P
Familiesa
Zb
P
Familiesa
Zb
P
PD1.1A72-0.2650.79142-0.6170.537460.1930.847
G720.2650.79146-0.1930.847420.6170.537
PD1.2A790.6400.52241-0.0800.936531.0010.317
G79-0.6400.52253-1.0010.317410.0800.936
Table 3 TDT of association with HBV infection-affection status in 539 trios selected from the HBV infectious nuclear families.
SNPFrequencyAlleleTrio TDT (n probands with allele)
Transmitted
Not transmitted
P
PD1.10.484A26300.689
0.516G30260.689
PD1.20.527A32331.000
0.473G33321.000

In addition, haplotype analysis showed that four haplotypes, PD-1.1 A/PD-1.2 A (46.3%), PD-1.1 G/PD-1.2 G (45.4%), PD-1.1 G/PD-1.2 A (4.7%) and PD-1.1 A/PD-1.2 G (3.5%), were reconstructed. FBAT was utilized in analyzing the data from 256 nuclear families. As shown in Table 4, the transmitted haplotypes were not associated with genetic susceptibility of chronic HBV infection in each of additive, dominant and recessive models (P > 0.05).

Table 4 The associations between PD-1 haplotypes and chronic HBV infection by family-based association test.
HaplotypeAdditive model
Dominant model
Recessive model
PD-1.1/1.2FamiliesaZbPFamiliesaZbPFamiliesaZbP
A/A70.80.3060.76048.8-0.6920.489351.2430.214
G/G66.50.0010.99945.5-0.1880.851320.2070.836
G/A20.20.2700.78719.20.3300.741---
A/G14.5-1.0900.27614.5-1.0900.276---
DISCUSSION

In humans, the molecular mechanisms of the association between HBV susceptibility and the PD-1 gene polymorphisms remain to be clarified. One possible mechanism is that selected PD-1 SNPs may be associated with an alteration in the level of expression of the PD-1 gene or possibly as a result of linkage disequilibrium with other PD-1 gene polymorphisms which differentially control PD-1 gene transcription. This possibility is supported by the identification of several SNPs in SLE, including in the promoter, introns, exon 5 and 3’-untranslated region of the PD-1 gene, and finding that the SNP PD-1.3 was involved in susceptibility to SLE in the European population while PD-1.1 and PD-1.2 was not[17]. However, the SNP PD-1.3 is not polymorphic in the Chinese population[20] and such discrepancies are worth further research. Another possible explanation is the connection between this SNP and the functional change in the PD-1 protein via linkage disequilibrium with other nucleotide polymorphisms that alter the sequence and structure of the PD-1 protein. This possibility may be tested by evaluating the ability of PD-1 protein to transduce inhibitory signals in response to ligand stimulation in individuals with different genotypes. However, it still cannot exclude the possibility that another genuine HBV susceptibility gene is located adjacent to the PD-1 gene that affects the association between the PD-1 gene and chronic HBV susceptibility. For instance, CTLA-4 gene was associated with chronic HBV infection[9,29] and the location of PD-1 is 2q37.3 in chromosome 2, almost nearer to CTLA-4 gene at 2q33. Therefore, based on the above considerations, our study employed a family-based association study in evaluating the genetic association of PD-1 to chronic HBV infection in Chinese patients; we suspected an immense role of PD-1 SNPs in chronic HBV infection.

However, in the present study, we have found no association of SNPs PD-1.1 A > G and PD-1.2 A > G in patients with chronic HBV infection. What is more, the major A or G allele from heterozygous parents did not transmit to affected offspring (P > 0.05). At the same time, we constructed four haplotypes comprising alleles of marker PD-1.1 G/A and PD-1.2 G/A and none of these haplotypes demonstrated an association (P > 0.05). In earlier studies, polymorphism of PD-1 SNPs (PD-1.1 and PD-1.2) in SLE, RA and T1D patients has been reported. Prokunina et al[17] reported PD-1.1 G > A and PD-1.2 G > A were not associated with SLE in European, Wang et al[19] validated their results in Taiwan Chinese. In addition, Asad et al[21] reported that PD-1.2 SNPs was not associated with T1D in Swedish. In these studies, no significant association was observed which is similar to our results. Conversely, Kong et al[20] reported that AA genotype of SNP PD-1.1 was associated with a decreased risk for developing RA in Hong Kong and that the PD-1.1 A allele can be considered a protective allele but the PD-1.1 G allele can not be called a risk allele by itself as it is a major allele in Caucasians (99%) and only a part of PD-1.1G-containing haplotypes are risk haplotypes. There may be several reasons for the differences between reported associations. For example, the underlying genetic factors involved in disease susceptibility might not be the same for chronic HBV infection as for RA or SLE[30-35]. The most likely reason for discrepancy between the other studies and ours is that our cohorts were larger than the Kong’s RA cohort which only consisted of 180 patients with RA in a case-control association study[20]. In addition, Kong’s study was based on case-control. Studies of case-control could lead to false-positive results because the control subjects were not ideally matched to the patients whereas the family-based study could avoid it by population stratification and reduced heterogeneity[36]. In some of the latest research, Zhang[37] and Zheng et al[38], investigated the association between chronic HBV infection and SNPs of the PD-1 gene and, although they used a large cohort of chronically HBV-infected patient samples and healthy controls, the results also indicated that neither PD-1.1 SNP nor P7209C > T, P8737A > G site in PD-1 gene was associated with the susceptibility of chronic HBV infection, validating our experimental results indirectly. Although Zhang et al[37] reported that PD-1.6 polymorphism may have a predisposing role in the disease progress, this needs further research. Therefore, the precision in estimating population frequencies among patients should be better in our investigation. Another reason for discrepancies is due to a large variation in the frequencies of PD-1 polymorphisms among different ethnic groups. For example, the PD-1.1 A allele is common in the Chinese population (49%) and in Mexican Indians (63%) while it is rare in Europeans (1%) and Africans (4%) (Prokunina-Olsson et al: unpublished observations); even James et al[34] reported PD-1.1 and PD-1.2 SNPs were not found in British Caucasians from southwest England. Moreover, it is known that complicated disorders like HBV depend on the interaction of multiple factors and no particular gene is uniquely responsible for the disease. Sometimes environmental factors may play a role in influencing susceptibility in different populations[39]. That could be an important reason why there are negative results in some research. For this reason, we cannot rule out the susceptibility of these genes even though the results are negative.

In conclusion, the present study has demonstrated that PD-1 gene polymorphism has no correlation with genetic susceptibility of chronic HBV infection in Chinese patients. Further genetic studies need to be replicated in populations of ancestries other than Chinese to estimate the polymorphisms and the information of the linkage disequilibrium (LD) between the other SNPs and the studied SNP.

COMMENTS
Background

Hepatitis B virus (HBV) infection remains a major public health problem. After infection, the diversity of the HBV disease spectrum and clinical course was attributed, to a very large extent, to the host immunological and genetic factors including single nucleotide polymorphisms (SNPs) of a variety of genes. Related gene polymorphism has important role in the understanding of the pathogenesis of disease and formulation of the prevention and treatment.

Research frontiers

In recent years, the association between the immune related gene polymorphism and HBV infection has been hot research, ranging from classic human leukocyte antigen (HLA), cytokines and some protein genes to a large study of genetic pathways and the whole genome scanning.

Innovations and breakthroughs

Recent reports have highlighted the importance of the PD-1 pathway in T-cell response in different stages of chronic HBV infection. This study firstly investigated the association between the polymorphisms of PD-1 (PD-1.1 and PD-1.2) and genetic susceptibility of chronic HBV infection by family-based association analysis method. The family based research excluded the effect of environment which could not be avoided in case-control design.

Applications

By discussing the association between the polymorphisms of PD-1 (PD-1.1 and PD-1.2) and genetic susceptibility of chronic HBV infection, this study may represent a future strategy for researching HBV infection by family-based association analysis method.

Terminology

PD-1: A 55 kDa transmembrane protein containing an immunological receptor tyrosine-based inhibitory motif which was originally isolated from a T-cell line exhibiting a high sensitivity to apoptosis. As a negative co-stimulatory receptor, PD-1 interacts with its ligands, PD-L1 and PD-L2, to attenuate T-cell, B-cell and APC-cell responses and appears to be particularly important for regulating immune cells tolerance.

Peer review

The manuscript described interesting data on the potential link between SNP in PD-1 and chronic HBV. As the data are contradictory to other reported findings, the authors are encouraged to analyze more carefully their experimental approach and interpret differences with others’ data objectively.

Footnotes

Peer reviewers: Ronald J Wong, Senior Research Scientist, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Dr, Rm S230, Stanford, CA, United States; Wei-Ning Chen, Associate Professor, School of Chemical and Biomedical Engineering, Nanyang Technological University 62, Nanyang Drive, N1.2, Singapore 637459, Singapore; Nattiya Hirankarn, MD, Associated Professor, Immunology Unit, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Rama 4 road, Bangkok 10330, Thailand

S- Editor Zhang HN L- Editor Roemmele A E- Editor Zhang L

References
1.  Luo KX Hepatitis B: Basic biology and clinical science. Second edition. Beijing: People’s Medical Publishing House 2001; 56-70.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Chu CJ, Lok AS. Clinical significance of hepatitis B virus genotypes. Hepatology. 2002;35:1274-1276.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Thursz M. Genetic susceptibility in chronic viral hepatitis. Antiviral Res. 2001;52:113-116.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Frodsham AJ. Host genetics and the outcome of hepatitis B viral infection. Transpl Immunol. 2005;14:183-186.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Lin TM, Chen CJ, Wu MM, Yang CS, Chen JS, Lin CC, Kwang TY, Hsu ST, Lin SY, Hsu LC. Hepatitis B virus markers in Chinese twins. Anticancer Res. 1989;9:737-741.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ramezani A, Hasanjani Roshan MR, Kalantar E, Eslamifar A, Banifazl M, Taeb J, Aghakhani A, Gachkar L, Velayati AA. Association of human leukocyte antigen polymorphism with outcomes of hepatitis B virus infection. J Gastroenterol Hepatol. 2008;23:1716-1721.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Gao QJ, Liu DW, Zhang SY, Jia M, Wu LH. Association between IFN-gamma+874 polymorphisms and the clinical outcomes of hepatitis B and/or hepatitis C virus infection. Zhonghua Liu Xing Bing Xue Za Zhi. 2010;31:324-328.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Kummee P, Tangkijvanich P, Poovorawan Y, Hirankarn N. Association of HLA-DRB1*13 and TNF-alpha gene polymorphisms with clearance of chronic hepatitis B infection and risk of hepatocellular carcinoma in Thai population. J Viral Hepat. 2007;14:841-848.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Thio CL, Mosbruger TL, Kaslow RA, Karp CL, Strathdee SA, Vlahov D, O’Brien SJ, Astemborski J, Thomas DL. Cytotoxic T-lymphocyte antigen 4 gene and recovery from hepatitis B virus infection. J Virol. 2004;78:11258-11262.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Deng G, Zhou G, Zhai Y, Li S, Li X, Li Y, Zhang R, Yao Z, Shen Y, Qiang B. Association of estrogen receptor alpha polymorphisms with susceptibility to chronic hepatitis B virus infection. Hepatology. 2004;40:318-326.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Zhou J, Lu L, Yuen MF, Lam TW, Chung CP, Lam CL, Zhang B, Wang S, Chen Y, Wu SH. Polymorphisms of type I interferon receptor 1 promoter and their effects on chronic hepatitis B virus infection. J Hepatol. 2007;46:198-205.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4:336-347.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci USA. 2001;98:13866-13871.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Peng G, Li S, Wu W, Tan X, Chen Y, Chen Z. PD-1 upregulation is associated with HBV-specific T cell dysfunction in chronic hepatitis B patients. Mol Immunol. 2008;45:963-970.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Zhang Z, Zhang JY, Wherry EJ, Jin B, Xu B, Zou ZS, Zhang SY, Li BS, Wang HF, Wu H. Dynamic programmed death 1 expression by virus-specific CD8 T cells correlates with the outcome of acute hepatitis B. Gastroenterology. 2008;134:1938-1949, 1949 e1931-1933.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Finger LR, Pu J, Wasserman R, Vibhakar R, Louie E, Hardy RR, Burrows PD, Billips LG. The human PD-1 gene: complete cDNA, genomic organization, and developmentally regulated expression in B cell progenitors. Gene. 1997;197:177-187.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Prokunina L, Castillejo-Lopez C, Oberg F, Gunnarsson I, Berg L, Magnusson V, Brookes AJ, Tentler D, Kristjansdottir H, Grondal G. A regulatory polymorphism in PDCD1 is associated with susceptibility to systemic lupus erythematosus in humans. Nat Genet. 2002;32:666-669.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Nielsen C, Laustrup H, Voss A, Junker P, Husby S, Lillevang ST. A putative regulatory polymorphism in PD-1 is associated with nephropathy in a population-based cohort of systemic lupus erythematosus patients. Lupus. 2004;13:510-516.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Wang SC, Chen YJ, Ou TT, Wu CC, Tsai WC, Liu HW, Yen JH. Programmed death-1 gene polymorphisms in patients with systemic lupus erythematosus in Taiwan. J Clin Immunol. 2006;26:506-151.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Kong EK, Prokunina-Olsson L, Wong WH, Lau CS, Chan TM, Alarcon-Riquelme M, Lau YL. A new haplotype of PDCD1 is associated with rheumatoid arthritis in Hong Kong Chinese. Arthritis Rheum. 2005;52:1058-1062.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Asad S, Nikamo P, Torn C, Landin-Olsson M, Lernmark A, Alarcon-Riquelme M, Kockum I. No evidence of association of the PDCD1 gene with Type 1 diabetes. Diabet Med. 2007;24:1473-1477.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Kroner A, Mehling M, Hemmer B, Rieckmann P, Toyka KV, Maurer M, Wiendl H. A PD-1 polymorphism is associated with disease progression in multiple sclerosis. Ann Neurol. 2005;58:50-57.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Liu X, Hu LH, Li YR, Chen FH, Ning Y, Yao QF. Programmed cell death 1 gene polymorphisms is associated with ankylosing spondylitis in Chinese Han population. Rheumatol Int. 2009;Dec 12.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Newby PR, Roberts-Davies EL, Brand OJ, Heward JM, Franklyn JA, Gough SC, Simmonds MJ. Tag SNP screening of the PDCD1 gene for association with Graves’ disease. Clin Endocrinol (Oxf). 2007;67:125-128.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Chinese Society of Infectious Diseases and Parasitology and Chinese Society of Hepatology of Chinese Medical Association. Chinese medicine national program for prevention and treatment ofviral hepatitis. Zhonghua Gan Zang Bing Za Zhi. 2000;8:324-325.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Rabinowitz D, Laird N. A unified approach to adjusting association tests for population admixture with arbitrary pedigree structure and arbitrary missing marker information. Hum Hered. 2000;50:211-223.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Horvath S, Xu X, Lake SL, Silverman EK, Weiss ST, Laird NM. Family-based tests for associating haplotypes with general phenotype data: application to asthma genetics. Genet Epidemiol. 2004;26:61-69.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Excoffier L, Slatkin M. Maximum-likelihood estimation of molecular haplotype frequencies in a diploid population. Mol Biol Evol. 1995;12:921-927.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Schott E, Witt H, Pascu M, van Boemmel F, Weich V, Bergk A, Halangk J, Muller T, Puhl G, Wiedenmann B. Association of CTLA4 single nucleotide polymorphisms with viral but not autoimmune liver disease. Eur J Gastroenterol Hepatol. 2007;19:947-951.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Abelson AK, Johansson CM, Kozyrev SV, Kristjansdottir H, Gunnarsson I, Svenungsson E, Jonsen A, Lima G, Scherbarth HR, Gamron S. No evidence of association between genetic variants of the PDCD1 ligands and SLE. Genes Immun. 2007;8:69-74.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Ferreiros-Vidal I, D’Alfonso S, Papasteriades C, Skopouli FN, Marchini M, Scorza R, Migliaresi S, Sebastiani GD, Endreffy E, Mavromati M. Bias in association studies of systemic lupus erythematosus susceptibility due to geographical variation in the frequency of a programmed cell death 1 polymorphism across Europe. Genes Immun. 2007;8:138-146.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Iwamoto T, Ikari K, Inoue E, Toyama Y, Hara M, Yamanaka H, Tomatsu T, Momohara S, Kamatani N. Failure to confirm association between PDCD1 polymorphisms and rheumatoid arthritis in a Japanese population. J Hum Genet. 2007;52:557-560.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Lin SC, Yen JH, Tsai JJ, Tsai WC, Ou TT, Liu HW, Chen CJ. Association of a programmed death 1 gene polymorphism with the development of rheumatoid arthritis, but not systemic lupus erythematosus. Arthritis Rheum. 2004;50:770-775.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  James ES, Harney S, Wordsworth BP, Cookson WO, Davis SJ, Moffatt MF. PDCD1: a tissue-specific susceptibility locus for inherited inflammatory disorders. Genes Immun. 2005;6:430-437.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Chen DQ, Zeng Y, Zhou J, Yang L, Jiang S, Huang JD, Lu L, Zheng BJ. Association of candidate susceptible loci with chronic infection with hepatitis B virus in a Chinese population. J Med Virol. 2010;82:371-378.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Glaser B, Holmans P. Comparison of methods for combining case-control and family-based association studies. Hum Hered. 2009;68:106-116.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Zhang G, Liu Z, Duan S, Han Q, Li Z, Lv Y, Chen J, Lou S, Li N. Association of polymorphisms of programmed cell death-1 gene with chronic hepatitis B virus infection. Hum Immunol. 2010;71:1209-1213.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Zheng L, Li D, Wang F, Wu H, Li X, Fu J, Chen X, Wang L, Liu Y, Wang S. Association Between Hepatitis B Viral Burden in Chronic Infection and a Functional Single Nucleotide Polymorphism of the PDCD1 Gene. J Clin Immunol. 2010;30:855-860.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Yashima Y, Oyama T, Pham TT, Yamaguchi T, Narai R, Isse T, Uramoto H, Sugio K, Yasumoto K, Kagawa N. Hazardous environmental factors enhance impairment of liver function in HBV and HCV hepatitis. Front Biosci. 2009;14:4071-4078.  [PubMed]  [DOI]  [Cited in This Article: ]