Next Article in Journal / Special Issue
Development of a Multiplex and Cost-Effective Genotype Test toward More Personalized Medicine for the Antiplatelet Drug Clopidogrel
Previous Article in Journal
Huperzine A Ameliorates Cognitive Deficits in Streptozotocin-Induced Diabetic Rats
Previous Article in Special Issue
Polymorphisms in DNA Repair Genes (APEX1, XPD, XRCC1 and XRCC3) and Risk of Preeclampsia in a Mexican Mestizo Population
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Different Role of Tumor Necrosis Factor-α Polymorphism in Non-Hodgkin Lymphomas among Caucasian and Asian Populations: A Meta-Analysis

Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2014, 15(5), 7684-7698; https://doi.org/10.3390/ijms15057684
Submission received: 12 March 2014 / Revised: 15 April 2014 / Accepted: 23 April 2014 / Published: 5 May 2014
(This article belongs to the Special Issue Human Single Nucleotide Polymorphisms and Disease Diagnostics)

Abstract

:
Tumor necrosis factor-α (TNF-α) is an immunoregulatory cytokine involved in B- and T-cell function, and also plays an important role in inflammation and cancer. TNF-α-308G>A has been associated with constitutively elevated TNF-α expression. Several studies have reported the association between the TNF-α-308G>A polymorphism and non-Hodgkin lymphomas (NHL) risk, however, results are still inconsistent. To solve these conflicts, we conducted the first meta-analysis to assess the effect of TNF-α-308G>A polymorphism on the risk of NHL and various subtypes (additive model) including 10,619 cases and 12,977 controls in Caucasian and Asian populations. Our meta-analysis indicated that TNF-α-308G>A polymorphism is not associated with NHL risk when pooling all studies together (OR = 1.06, 95% CI: 0.92–1.23, p = 0.413). In stratified analyses, we found TNF-α-308A allele was significantly associated with higher risk of NHL, B-cell lymphomas (BCL), T-cell lymphomas (TCL) and diffuse large B-cell lymphomas (DLBCL) in Caucasians (OR = 1.22, 95% CI: 1.06–1.40, p = 0.007; OR = 1.18, 95% CI: 1.03–1.34, p = 0.014; OR = 1.20, 95% CI: 1.01–1.42, p = 0.040; OR = 1.21, 95% CI: 1.11–1.32, p < 0.001, respectively). Interestingly, it was associated with decreased risk of NHL, BCL and DLBCL in Asians (OR = 0.75, 95% CI: 0.66–0.86, p < 0.001; OR = 0.70, 95% CI: 0.52–0.94, p = 0.018; OR = 0.70, 95% CI: 0.57–0.86, p = 0.001). These findings also suggest TNF-α might play a distinct role in pathogenesis of NHL in different populations.

1. Introduction

Non-Hodgkin lymphomas (NHL), a complex group of heterogeneous diseases of uncontrolled B- or T-cell proliferation with distinct clinical and histological features, accounts for approximately 90% of all malignancy lymphomas [1]. Malignant transformation of B- or T-cells can occur at different stages of maturation, which reflects the heterogeneity of malignancies with various biologic and clinical behaviors. B-cell lymphomas (BCL) comprise 90% of NHL. Diffuse large B-cell lymphomas (DLBCL) and follicular lymphomas (FL) are the two major subtypes of BCL. Clinical outcome of NHL varies from subtype, diagnosis and response to treatment, however, prognosis of T-cell lymphoma (TCL) is usually worse than that of BCL. Etiology of NHL is still poorly understood, although epidemiological studies have shown that individuals with innate or acquired immune deficiencies, immunosuppression and infection are at increased risk of NHL [2,3]. Recently, accumulating evidence has suggested that genetic variations such as single nucleotide polymorphisms (SNPs) are associated with NHL risk and survival [49]. Moreover, previous studies showing a 2- to 3-fold risk of NHL with a family history of hematological malignancies indicates that genetic factors might play a critical role in NHL pathogenesis [1012].
Tumor necrosis factor-α (TNF-α) is one of the most important pro-inflammatory and tumor-related cytokines for its regulating immune response, inflammation, Th1/Th2 balance and lymphomagenesis [13]. Increased serum values of TNF-α have been detected in autoimmune disease and many malignancies including lymphomas [1417]. TNF-α-308G>A (rs1800629) SNP has increased susceptibility to many kinds of tumors and autoimmune diseases, such as hepatocellular carcinoma, myeloma, lymphoma, ulcerative colitis, and Crohn’s disease [1820]. TNF-α-308A allele is associated with higher constitutive and inducible TNF-α expression by affecting a consensus binding site of a transcription factor named activator protein-2 (AP-2) [21,22]. Studies using knockout mouse have supported that this cytokine could affect progression of BCL directly or indirectly [23,24]. Although the TNF-α-308G>A polymorphism has been widely assessed in association with NHL in different ethnicities, due to various sample sizes and genotyping methods, possibly because of NHL heterogeneity and other reasons, the results are still controversial.
In this study, we conducted the first comprehensive meta-analysis to test whether the TNF-α-308 polymorphism is associated with NHL overall risk or its subtypes, especially BCL, TCL, DLBCL, FL, chronic lymphocytic leukemia/small lymphocytic lymphomas (CLL/SLL), mantel cell lymphomas (MCL), mucosal-associated lymphomas (MALT), peripheral T-cell lymphomas (PTCL) and natural killer/T-cell lymphomas (NK/TCL). We also performed subgroup analysis by descent (Caucasians and Asians) to assess a possible factor that might influence the overall results. Therefore, this study might have more statistical power and increase precision to estimate association between TNF-α-308 polymorphism and its effect on NHL.

2. Results and Discussion

2.1. Eligible Studies

In the initial screening for key words, 405 potential articles were identified in PubMed, Embase and Cochrane Library. After removing duplication, 321 articles were needed for further assessment. Among them, 293 were excluded because of inappropriate study design or control samples. Of the remaining 28 relevant articles, 8 articles were excluded for using the same patients. 2 articles were also excluded for their controls in concordance with Hardy-Weinberg equilibrium (HWE). With strict including criteria, the final pool of eligible articles consisted of 18 articles involving a total of 10,619 patients with NHL and 12,977 healthy controls. Because of the large sample size, Caucasians and Asians were considered as population stratification in this meta-analysis. Table 1 shows characteristics of eligible articles including ethnicity, genotyping method, number of cases and controls and NHL pathological types. In fact, at the primary data extraction, allele frequencies in all controls of one study, which performed by Skibola, did not fulfill HWE [25]. This study is a meta- and pooled analysis adding more genotyping data to the initial pooled report [26] to confirm the association between TNF/LTA polymorphism and NHL risk in Caucasian and Asian populations. For the new subjects (including Caucasians and Asians) not included in the initial report (all were Caucasians) [26], allele frequency of TNF-α-308G>A in controls met HWE (p = 0.916). But TNF-α-308G>A in all controls in the initial report was not consistent with HWE (p = 0.0007). We analyzed the initial report composed of 8 subgroups comprehensively [26]. Finally, we excluded data of EPILYMPH-Spain, University of California San Francisco and the NCI-SEER Seattle subgroup, in which controls did not fulfill HWE, and extracted data successfully. Since Skibola et al. [25] conducted this large pooled analysis in TNF polymorphism on NHL risk in Caucasians and Asians, we separated this paper into two studies according to population. In addition, 13 studies were conducted on Caucasians [16,2738], and 4 were on Asians [3942]. Several genotyping methods were used, including allelic specific polymerase chain reaction (ASPCR), polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), polymerase chain reaction-solid-phase minisequencing (PCR-SPM), polymerase chain reaction-ligation detection reaction (PCR-LDR), TaqMan, Sequenom and sequencing.

2.2. Quantitative Synthesis

Based on a large pooled sample size, we analyzed TNF-α-308G>A polymorphism effects on risks of NHL, BCL, TCL and subtypes (DLBCL, FL, CLL/SLL, MCL, MALT, PTCL and NK/TCL) in additive model (A vs. G) which stratified by ethnicity (Caucasians and Asians). Results of meta-analysis and primary data extracted from studies are listed in Tables 2 and 3.

2.2.1. TNF-α-308G>A and NHL

Of the combined 10,619 patients with NHL and 12,977 controls, no risk association was observed in TNF-308G>A polymorphism and NHL with significant heterogeneity between studies (OR = 1.06, 95% CI: 0.92–1.23, p = 0.413; I2 = 75.0%, phet < 0.001). In subgroup analysis based on population, significant associations were detected in Caucasian and Asian populations, respectively. In Caucasians with 7893 cases and 8447 controls, participants with TNF-308A allele had an increased NHL risk (OR = 1.22, 95% CI: 1.06–1.40, p = 0.007; I2 = 60.7%, phet = 0.002). However, in Asians with 2726 cases and 4530 controls, decreased risk was observed (OR = 0.75, 95% CI: 0.66–0.86, p < 0.001; I2 = 0.0%, phet = 0.670). Figure 1 shows the forest plot of the overall association between TNF-α-308G>A polymorphism and NHL risk in additive model (A vs. G) stratified by ethnicity.
To evaluate the influence of each study on the pooled ORs in two subgroups, we deleted single study at a time to recalculate the influence of individual study for the outcome of the meta-analysis. The pooled ORs were stable and in an effective interval with statistical significant though the fixed-effect in additive model estimating before or after any single study deleted in each group (data not shown). These indicated that the results of this meta-analysis were reliable and had not been overly influenced by any one of studies.
We performed Begg’s funnel plot and Egger’s test to evaluate the publication bias of all included studies. Figure 2 shows no evidence of obvious asymmetry in overall analysis for TNF-α-308G>A polymorphism in additive model (pBegg’s = 0.529). Egger’s test also suggested no significant publication bias existed in this meta-analysis (additive model, p = 0.780).

2.2.2. TNF-α-308G>A and B- or T-CL

Thirteen studies comprising a total of 20,064 participants (8092 cases with BCL and 11,972 controls) and 4 studies including 11,832 participants (1100 cases with TCL and 10,732 controls) were analyzed for an association between TNF-α-308G>A polymorphism and BCL or TCL risk.
For BCL analysis, the pooled OR across all studies was not statistically significant (OR = 1.07, 95% CI: 0.91–1.26, p = 0.411; I2 = 74.4%, phet < 0.001). Increased risk was found in Caucasians (OR = 1.18, 95% CI: 1.03–1.34, p = 0.014; I2 = 44.6%, phet = 0.054), decreased risk was detected in Asians (OR = 0.70, 95% CI: 0.52–0.94, p = 0.018; I2 = 41.9%, phet = 0.189). In TCL analysis, TNF-308A associated with a higher NHL risk in Caucasians (OR = 1.20, 95% CI: 1.01–1.42, p = 0.040; I2 = 0.0%, phet = 0.361).

2.2.3. TNF-α-308G>A and NHL Subtypes

For DLBCL, the most common NHL subtype, there were 3353 cases and 10,817 controls included in the analysis. Consistent with the results for all NHL, the TNF-α-308A allele was associated with an increased risk of DLBCL in Caucasians (OR = 1.21, 95% CI: 1.11–1.32, p < 0.001; I2 = 0.0%, phet = 0.491), but with a decreased risk in Asians (OR = 0.70, 95% CI: 0.57–0.86, p = 0.001; I2 = 0.0%, phet = 0.908). The pooled OR was 0.97 (95% CI: 0.75–1.26, p = 0.840; I2 = 83.9%, phet < 0.001).
No associations were found in TNF-α-308G>A polymorphism with FL, CLL/SLL, MCL, MALT, PTCL and NK/TCL in overall and each ethnic subgroup.

2.3. Discussion

In the pooled analysis of 18 articles, we found TNF-α-308G>A polymorphism to be significantly associated with NHL risk in Caucasians and Asians. We provided evidence that subjects with TNF-α-308A allele had an increased risk of NHL in Caucasians, and had a decreased risk in Asians. Similar results were confirmed in analyses of BCL and DLBCL. Further, the TNF-α-308A allele was positively associated with risks of TCL in Caucasians. Our study highlights the effect of TNF-α gene polymorphism on risks of NHL and its subtypes in different populations. These findings indicate a potential connection between constitutively higher TNF-α expression and pathogenesis of NHL.
TNF-α is a transmembrane protein and mainly produced by macrophages and is expressed at low levels in a wide variety of cells. TNF-α mediates its effects through TNF-α receptor 1 and 2 (TNFR1 and TNFR2) by ligand passing and signal transduction. TNFR1 has a death domain that could interact with TNF-α receptor-associated death domain (TRADD), sequentially recruiting proteins to induce caspase-3 activation for apoptosis. TRADD could also bind TNF receptor-associated factor 2 (TRAF2) to recruits proteins activating IKK, GCK, and RIP, which finally leads to the NF-κB, JNK, and MAPK pathway activation for anti-apoptosis and cell survival. Although TNFR2 lacks the death domain, it could also bind TRAF2 to active an anti-apoptosis pathway [13,43]. Aberrant NF-κB activation is a hallmark of several lymphomas for promoting continuous lymphocyte proliferation, which is also directly linked to disease promotion [44,45]. When cells are exposed to TNF-α, NF-κB pathway activation leads to the expression of many genes to cause chronic inflammation, which stimulates tumor growth. Dysregulated TNF-α contributes directly to the transformed state in many cancers, especially those of BCL [46]. Collectively, TNF-α acting as an immunoregulatory cytokine builds a bridge between inflammation and cancer by activating many biological pathways including the nuclear factor-κB (NF-κB) pathway in promoting cell proliferation, survival, transformation, invasion and angiogenesis.
Previous studies suggest higher expression of TNF-α is associated with NHL risk at the time of diagnosis [1417]. These studies do not contradict the results of TNF-α-308A allele inducing higher constitutively TNF-α expression associated with decreased NHL risk in Asians. With a heterogeneous malignancy and population diversity, we believe the level of constitutively TNF-α expression must play a different vital role at the step of NHL initiation in Caucasians and Asians, although the reasons for this have not been understood. Once a tumor forms, it secretes TNF-α to promote its survival, proliferation and metastasis. A subtype of TCL failing to express TNF-α and frequently with the TNF-α gene promoter methylated [47] indicates that epigenetic changes might also influence NHL susceptibility together. Environmental, occupational exposure and pathogenic agent infection (such as Epstein-Barr virus and human T-cell leukemia virus-1) are the well-known risk factors for NHL [48,49]. Therefore, genetic, epigenetic, tumor microenvironment, environment and their interaction could together contribute to NHL progression. Few studies about this kind of interaction relative to NHL susceptibility have been published. Due to insufficient data, our meta-analysis did not combine the effects of these factors in an association analysis between genetic variation and NHL risk. Much more precise investigations should be performed to clarify the true association of these types of interactions with polymorphism and NHL.
TNF-α and LT-α gene lie in the major histocompatibility complex class III, telemetric to the class II and centrometric to class I gene. Therefore, TNF-α being in linkage disequilibrium (LD) with these genes may also be linked to another region, haplotype or extended, that can influence NHL development [50]. This meta-analysis only evaluated one SNP in the TNF-α gene, though it was not possible to analyze haplotypes with the present data. Further studies will be needed to pool data and analyze whether haplotypes comprising TNF-α-308G>A and other SNPs are linked to NHL risk, and clarify their function concomitantly.
Because of relatively low incidence of NHL, sample sizes of some studies included in this analysis are very small. The major strength of this study is the larger pooled sample size involving a total of 10,619 patients with NHL and 12,977 healthy controls for association study, which would largely minimize the possibility of chance findings. In addition, patients in our study were all Caucasians or Asians, which would exclude the biased results due to population stratification.
In conclusion, we performed the first comprehensive meta-analysis involving 10,619 patients with NHL and 12,977 controls from 18 articles to evaluate the association between TNF-α-308G>A polymorphism and NHL risk. Our study showed that TNF-α-308G>A SNP in the promoter region of TNF-α gene is associated with NHL risk. In addition, TNF-α-308A increases risks of NHL, BCL, TCL and DLBCL in the Caucasian population; however, interestingly, it reduces risks of NHL, BCL and DLBCL in the Asian population. This association might be mediated by constitutive changes of TNF-α expression in individuals carrying the -308A allele, to induce inflammatory responses or the alternative pathway which be involved in NHL initiation and progression. Our meta-analysis emphasizes that genetic variation plays a crucial role in cancer; theTNF-α-308G>A polymorphism might play a role in a specific subtype of NHL and its importance varies in different populations. Further studies should focus on the function of how variants affect NHL in different populations and the elucidation of the pathway involved which may eventually lead to a better understanding of tumorigenesis and contribute to the prevention of NHL.

3. Experimental Section

3.1. Publication Search

We carried out a search in three electronic databases PubMed, Embase and Cochrane Library to find relevant publications up to November 2013, using key words related to the TNF-α gene polymorphism in combination with various NHL subtypes [51]. The search was limited to studies that had been conducted on human subjects and without language restriction. Reference lists of the retrieved articles, reviews and editorials were also screened to find all additional eligible studies.

3.2. Inclusion Criteria

Selection of studies had to meet the following criteria: (1) case-control studies, family or sibling pairs studies were excluded; (2) published in English; (3) subjects were limited to adult and without autoimmune diseases, studies with children were also excluded; (4) DNA was extracted from peripheral blood leukocytes; (5) study described the association between TNF-α-308 polymorphism and NHL risk; (6) sufficient data for estimating odds ratio (OR) and its corresponding 95% confidence interval (95% CI); (7) control group fulfilled HWE. When the same subject group occurred in more than one study, only the complete study was chosen to be included in this meta-analysis.

3.3. Data Extraction

An initial screening of title and abstract was performed for the first step, followed by further screening based on full-text review. Information was independently extracted from all eligible publications by two investigators (K.Z. and J.D.), including the first author, publication year, ethnicity, sample size, genotyping method, the number of each genotype in cases and controls. For studies with subjects of different ethnic groups and had sufficient information, we extracted data separately for each ethnicity. Disagreements were resolved through discussion.

3.4. Statistical Analysis

We assessed the association between TNF-α-308G>A polymorphism and NHL risk by crude ORs and 95% CIs in an additive model. Heterogeneity among studies was examined with I2 statistics. In this meta-analysis, I2 > 50% was defined as heterogeneity. Fixed-effect model (Mantel-Haenszel method) was used to evaluate inter-study heterogeneity. If heterogeneity existed, random-effect model (DerSimonian-Laird method) was used. Z test was used to determine the pooled OR and 95% CI. Analyses were also conducted on the subgroups of studies based on ethnicity. The potential influence of publication bias was assessed using Begg’s funnel plot and Egger’s linear regression test [52,53]. To evaluate the effect of one single study on overall risk of NHL, sensitivity analyses by excluding every study and recalculating ORs and 95% CI were conducted [54]. HWE in controls of each study was examined by the Pearson’s goodness-of-fit χ2 test. All statistical tests were carried out with SPSS 16.0 (SPSS Inc., Chicago, IL, USA) and Stata 12.0 (StataCorp, College Station, TX, USA). A 2-tailed p < 0.05 was considered as statistical significance.

4. Conclusions

We performed the first comprehensive meta-analysis involving 10,619 patients with NHL and 12,977 controls from 18 articles to evaluate the association between TNF-α-308G>A polymorphism and NHL risk. Our study showed that TNF-α-308G>A SNP in the promoter region of TNF-α gene is associated with NHL risk. In addition, TNF-α-308A increases risks of NHL, BCL, TCL and DLBCL in the Caucasian population; interestingly, this polymorhism reduces risks of NHL, BCL and DLBCL in the Asian population. This association might be mediated by constitutive changes of TNF-α expression in individuals carrying the -308A allele, to induce inflammatory responses or the alternative pathway which be involved in NHL initiation and progression. Our meta-analysis emphasizes that genetic variation plays a crucial role in cancer; the TNF-α-308G>A polymorphism might play a role in a specific subtype of NHL and its importance may vary in different populations. Further studies should be focused on how variants affect NHL in different populations and the elucidation of the pathways involved which may eventually lead to a better understanding of tumorigenesis and contribute to the prevention of NHL.

Acknowledgments

This work was supported by National Natural Science Foundation of China (No. 81302052) and Natural Science Foundation of Beijing (No. 7142061).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

TNF
tumor necrosis factor
NHL
non-Hodgkin lymphomas
BCL
B-cell lymphomas
TCL
T-cell lymphomas
DLBCL
diffuse large B-cell lymphomas
FL
follicular lymphomas
CLL/SLL
chronic lymphocytic leukemias/small lymphocytic lymphomas
MCL
mantel cell lymphomas
MALT
mucosal-associated lymphomas
PTCL
peripheral T-cell lymphomas
NK/TCL
natural killer/T-cell lymphomas
OR
odds ratio
95% CI
95% confidence interval
  • Author ContributionsConceived and designed the experiments: K.Z. Performed the experiments: K.Z. and J.D. Analyzed the data: K.Z., J.D. and Y.Z. Wrote the paper: K.Z. J.D. and Y.Z.

References

  1. Shankland, K.R.; Armitage, J.O.; Hancock, B.W. Non-Hodgkin lymphoma. Lancet 2012, 380, 848–857. [Google Scholar]
  2. Evans, L.S.; Hancock, B.W. Non-Hodgkin lymphoma. Lancet 2003, 362, 139–146. [Google Scholar]
  3. Alexander, D.D.; Mink, P.J.; Adami, H.O.; Chang, E.T.; Cole, P.; Mandel, J.S.; Trichopoulos, D. The non-Hodgkin lymphomas: A review of the epidemiologic literature. Int. J. Cancer 2007, 120 Suppl 12, 1–39. [Google Scholar]
  4. Kumar, V.; Matsuo, K.; Takahashi, A.; Hosono, N.; Tsunoda, T.; Kamatani, N.; Kong, S.Y.; Nakagawa, H.; Cui, R.; Tanikawa, C.; et al. Common variants on 14q32 and 13q12 are associated with DLBCL susceptibility. J. Hum. Genet 2011, 56, 436–439. [Google Scholar]
  5. Skibola, C.F.; Conde, L.; Foo, J.N.; Riby, J.; Humphreys, K.; Sille, F.C.; Darabi, H.; Sanchez, S.; Hjalgrim, H.; Liu, J.; et al. A meta-analysis of genome-wide association studies of follicular lymphoma. BMC Genomics 2012, 13, 516. [Google Scholar]
  6. Zhai, K.; Tian, X.; Wu, C.; Lu, N.; Chang, J.; Huang, L.; Zhang, T.; Zhou, Y.; Qiao, Y.; Yu, D.; et al. Cytokine BAFF gene variation is associated with survival of patients with T-cell lymphomas. Clin. Cancer Res 2012, 18, 2250–2256. [Google Scholar]
  7. Berndt, S.I.; Skibola, C.F.; Joseph, V.; Camp, N.J.; Nieters, A.; Wang, Z.; Cozen, W.; Monnereau, A.; Wang, S.S.; Kelly, R.S.; et al. Genome-wide association study identifies multiple risk loci for chronic lymphocytic leukemia. Nat. Genet 2013, 45, 868–876. [Google Scholar]
  8. Tan, D.E.; Foo, J.N.; Bei, J.X.; Chang, J.; Peng, R.; Zheng, X.; Wei, L.; Huang, Y.; Lim, W.Y.; Li, J.; et al. Genome-wide association study of B cell non-Hodgkin lymphoma identifies 3q27 as a susceptibility locus in the Chinese population. Nat. Genet 2013, 45, 804–807. [Google Scholar]
  9. Vijai, J.; Kirchhoff, T.; Schrader, K.A.; Brown, J.; Dutra-Clarke, A.V.; Manschreck, C.; Hansen, N.; Rau-Murthy, R.; Sarrel, K.; Przybylo, J.; et al. Susceptibility loci associated with specific and shared subtypes of lymphoid malignancies. PLoS Genet 2013, 9, e1003220. [Google Scholar]
  10. Zhu, K.; Levine, R.S.; Gu, Y.; Brann, E.A.; Hall, I.; Caplan, L.S.; Baum, M.K. Non-Hodgkin’s lymphoma and family history of malignant tumors in a case-control study (United States). Cancer Causes Control 1998, 9, 77–82. [Google Scholar]
  11. Altieri, A.; Bermejo, J.L.; Hemminki, K. Familial risk for non-Hodgkin lymphoma and other lymphoproliferative malignancies by histopathologic subtype: The Swedish Family-Cancer Database. Blood 2005, 106, 668–672. [Google Scholar]
  12. Wang, S.S.; Slager, S.L.; Brennan, P.; Holly, E.A.; de Sanjose, S.; Bernstein, L.; Boffetta, P.; Cerhan, J.R.; Maynadie, M.; Spinelli, J.J.; et al. Family history of hematopoietic malignancies and risk of non-Hodgkin lymphoma (NHL): A pooled analysis of 10,211 cases and 11,905 controls from the International Lymphoma Epidemiology Consortium (InterLymph). Blood 2007, 109, 3479–3488. [Google Scholar]
  13. Sethi, G.; Sung, B.; Aggarwal, B.B. TNF: A master switch for inflammation to cancer. Front. Biosci 2008, 13, 5094–5107. [Google Scholar]
  14. Macia, J.; Gomez, X.; Esquerda, A.; Perez, B.; Callao, V.; Marzo, C. Value of the determination of TNF-alpha in the plasma of patients with non-Hodgkins lymphoma. Leuk. Lymphoma 1996, 20, 481–486. [Google Scholar]
  15. Adami, F.; Guarini, A.; Pini, M.; Siviero, F.; Sancetta, R.; Massaia, M.; Trentin, L.; Foa, R.; Semenzato, G. Serum levels of tumour necrosis factor-alpha in patients with B-cell chronic lymphocytic leukaemia. Eur. J. Cancer 1994, 30A, 1259–1263. [Google Scholar]
  16. Warzocha, K.; Ribeiro, P.; Bienvenu, J.; Roy, P.; Charlot, C.; Rigal, D.; Coiffier, B.; Salles, G. Genetic polymorphisms in the tumor necrosis factor locus influence non-Hodgkin’s lymphoma outcome. Blood 1998, 91, 3574–3581. [Google Scholar]
  17. Halida, Y.; Guo, X.H.; Aliya, R. Expression of plasma TNF-alpha and TNF-beta in different subtypes lymphoma and its significance. Zhonghua Xue Ye Xue Za Zhi 2011, 32, 695–697. (In Chinese) [Google Scholar]
  18. Wilson, A.G.; di Giovine, F.S.; Duff, G.W. Genetics of tumour necrosis factor-alpha in autoimmune, infectious, and neoplastic diseases. J. Inflamm 1995, 45, 1–12. [Google Scholar]
  19. Neben, K.; Mytilineos, J.; Moehler, T.M.; Preiss, A.; Kraemer, A.; Ho, A.D.; Opelz, G.; Goldschmidt, H. Polymorphisms of the tumor necrosis factor-alpha gene promoter predict for outcome after thalidomide therapy in relapsed and refractory multiple myeloma. Blood 2002, 100, 2263–2265. [Google Scholar]
  20. Ho, S.Y.; Wang, Y.J.; Chen, H.L.; Chen, C.H.; Chang, C.J.; Wang, P.J.; Chen, H.H.; Guo, H.R. Increased risk of developing hepatocellular carcinoma associated with carriage of the TNF2 allele of the -308 tumor necrosis factor-alpha promoter gene. Cancer Causes Control 2004, 15, 657–663. [Google Scholar]
  21. Kroeger, K.M.; Carville, K.S.; Abraham, L.J. The -308 tumor necrosis factor-alpha promoter polymorphism effects transcription. Mol. Immunol 1997, 34, 391–399. [Google Scholar]
  22. Wilson, A.G.; Symons, J.A.; McDowell, T.L.; McDevitt, H.O.; Duff, G.W. Effects of a polymorphism in the human tumor necrosis factor alpha promoter on transcriptional activation. Proc. Natl. Acad. Sci. USA 1997, 94, 3195–3199. [Google Scholar]
  23. Pikarsky, E.; Porat, R.M.; Stein, I.; Abramovitch, R.; Amit, S.; Kasem, S.; Gutkovich-Pyest, E.; Urieli-Shoval, S.; Galun, E.; Ben-Neriah, Y. NF-κB functions as a tumour promoter in inflammation-associated cancer. Nature 2004, 431, 461–466. [Google Scholar]
  24. Batten, M.; Fletcher, C.; Ng, L.G.; Groom, J.; Wheway, J.; Laabi, Y.; Xin, X.; Schneider, P.; Tschopp, J.; Mackay, C.R.; et al. TNF deficiency fails to protect BAFF transgenic mice against autoimmunity and reveals a predisposition to B cell lymphoma. J. Immunol 2004, 172, 812–822. [Google Scholar]
  25. Skibola, C.F.; Bracci, P.M.; Nieters, A.; Brooks-Wilson, A.; de Sanjose, S.; Hughes, A.M.; Cerhan, J.R.; Skibola, D.R.; Purdue, M.; Kane, E.; et al. Tumor necrosis factor (TNF) and lymphotoxin-alpha (LTA) polymorphisms and risk of non-Hodgkin lymphoma in the InterLymph Consortium. Am. J. Epidemiol 2010, 171, 267–276. [Google Scholar]
  26. Rothman, N.; Skibola, C.F.; Wang, S.S.; Morgan, G.; Lan, Q.; Smith, M.T.; Spinelli, J.J.; Willett, E.; de Sanjose, S.; Cocco, P.; et al. Genetic variation in TNF and IL10 and risk of non-Hodgkin lymphoma: A report from the InterLymph Consortium. Lancet Oncol 2006, 7, 27–38. [Google Scholar]
  27. Chouchane, L.; Ahmed, S.B.; Baccouche, S.; Remadi, S. Polymorphism in the tumor necrosis factor-alpha promotor region and in the heat shock protein 70 genes associated with malignant tumors. Cancer 1997, 80, 1489–1496. [Google Scholar]
  28. Demeter, J.; Porzsolt, F.; Ramisch, S.; Schmidt, D.; Schmid, M.; Messer, G. Polymorphism of the tumour necrosis factor-α and lymphotoxin-α genes in chronic lymphocytic leukaemia. Br. J. Haematol 1997, 97, 107–112. [Google Scholar]
  29. Fitzgibbon, J.; Grenzelias, D.; Matthews, J.; Lister, T.A.; Gupta, R.K. Tumour necrosis factor polymorphisms and susceptibility to follicular lymphoma. Br. J. Haematol 1999, 107, 388–391. [Google Scholar]
  30. Wihlborg, C.; Sjoberg, J.; Intaglietta, M.; Axdorph, U.; Pisa, E.K.; Pisa, P. Tumour necrosis factor-alpha cytokine promoter gene polymorphism in Hodgkin’s disease and chronic lymphocytic leukaemia. Br. J. Haematol 1999, 104, 346–349. [Google Scholar]
  31. Mainou-Fowler, T.; Dickinson, A.M.; Taylor, P.R.; Mounter, P.; Jack, F.; Proctor, S.J.; Nordon, J.; Middleton, P.G. Tumour necrosis factor gene polymorphisms in lymphoproliferative disease. Leuk. Lymphoma 2000, 38, 547–552. [Google Scholar]
  32. Juszczynski, P.; Kalinka, E.; Bienvenu, J.; Woszczek, G.; Borowiec, M.; Robak, T.; Kowalski, M.; Lech-Maranda, E.; Baseggio, L.; Coiffier, B.; et al. Human leukocyte antigens class II and tumor necrosis factor genetic polymorphisms are independent predictors of non-Hodgkin lymphoma outcome. Blood 2002, 100, 3037–3040. [Google Scholar]
  33. Hellmig, S.; Fischbach, W.; Goebeler-Kolve, M.E.; Folsch, U.R.; Hampe, J.; Schreiber, S. A functional promotor polymorphism of TNF-α is associated with primary gastric B-Cell lymphoma. Am. J. Gastroenterol 2005, 100, 2644–2649. [Google Scholar]
  34. Bel Hadj Jrad, B.; Chatti, A.; Laatiri, A.; Ahmed, S.B.; Romdhane, A.; Ajimi, S.; Chouchane, L. Tumor necrosis factor promoter gene polymorphism associated with increased susceptibility to non-Hodgkin’s lymphomas. Eur. J. Haematol 2007, 78, 117–122. [Google Scholar]
  35. Jevtovic-Stoimenov, T.; Kocic, G.; Pavlovic, D.; Macukanovic-Golubovic, L.; Marjanovic, G.; Djordjevic, V.; Tosic, N.; Pavlovic, S. Polymorphisms of tumor-necrosis factor-α-308 and lymphotoxin-α + 250: Possible modulation of susceptibility to apoptosis in chronic lymphocytic leukemia and non-Hodgkin lymphoma mononuclear cells. Leuk. Lymphoma 2008, 49, 2163–2169. [Google Scholar]
  36. Fernberg, P.; Chang, E.T.; Duvefelt, K.; Hjalgrim, H.; Eloranta, S.; Sorensen, K.M.; Porwit, A.; Humphreys, K.; Melbye, M.; Ekstrom Smedby, K. Genetic variation in chromosomal translocation breakpoint and immune function genes and risk of non-Hodgkin lymphoma. Cancer Causes Control 2010, 21, 759–769. [Google Scholar]
  37. Ibrahim, A.; Abdel Rahman, H.; Khorshied, M.; Sami, R.; Nasr, N.; Khorshid, O. Tumor necrosis factor α-308 and Lymphotoxin α + 252 genetic polymorphisms and the susceptibility to non-Hodgkin lymphoma in Egypt. Leuk. Res 2012, 36, 694–698. [Google Scholar]
  38. Lech-Maranda, E.; Mlynarski, W.; Grzybowska-Izydorczyk, O.; Borowiec, M.; Pastorczak, A.; Cebula-Obrzut, B.; Klimkiewicz-Wojciechowska, G.; Wcislo, M.; Majewski, M.; Kotkowska, A.; et al. Polymorphisms of TNF and IL-10 genes and clinical outcome of patients with chronic lymphocytic leukemia. Genes Chromosom. Cancer 2013, 52, 287–296. [Google Scholar]
  39. Xiao, H.; Zhang, K. Genetic polymorphisms of tumor necrosis factor-α and lymphotoxin-α in Chinese patients with non-Hodgkin lymphoma. Ann. Hematol 2011, 90, 725–727. [Google Scholar]
  40. Hosgood, H.D., 3rd; Au, W.Y.; Kim, H.N.; Liu, J.; Hu, W.; Tse, J.; Song, B.; Wong, K.F.; Lee, J.J.; Chanock, S.J.; et al. IL10 and TNF variants and risk of non-Hodgkin lymphoma among three Asian populations. Int. J. Hematol 2013, 97, 793–799. [Google Scholar]
  41. Liu, J.; Song, B.; Wang, T.; Liu, Y.; Hao, J.; Yu, J. Genetic variations in CTLA-4, TNF-α, and LTA and susceptibility to T-cell lymphoma in a Chinese population. Cancer Epidemiol 2013, 37, 930–934. [Google Scholar]
  42. Nasiri, H.; Farajnia, S.; Rezamand, A.; Movassaghpour, A.A.; Esmaeili, H.A.; Monfaredan, A.; Mobarra, N.; Rahimifar, N.; Sahebi, L.; Farshdousti Hagh, M. Genetic variations of tumor necrosis factor-α-308 and lymphtoxin-α + 252 in non-hodgkin lymphoma and acute lymphoblastic leukemia patients. Iran. J. Basic Med. Sci 2013, 16, 990–995. [Google Scholar]
  43. Chu, W.M. Tumor necrosis factor. Cancer Lett 2013, 328, 222–225. [Google Scholar]
  44. Jost, P.J.; Ruland, J. Aberrant NF-κB signaling in lymphoma: Mechanisms, consequences, and therapeutic implications. Blood 2007, 109, 2700–2707. [Google Scholar]
  45. Shih, V.F.; Tsui, R.; Caldwell, A.; Hoffmann, A. A single NF κB system for both canonical and non-canonical signaling. Cell Res 2011, 21, 86–102. [Google Scholar]
  46. Younes, A.; Aggarwall, B.B. Clinical implications of the tumor necrosis factor family in benign and malignant hematologic disorders. Cancer 2003, 98, 458–467. [Google Scholar]
  47. Zhang, Q.; Wang, H.Y.; Bhutani, G.; Liu, X.; Paessler, M.; Tobias, J.W.; Baldwin, D.; Swaminathan, K.; Milone, M.C.; Wasik, M.A. Lack of TNFalpha expression protects anaplastic lymphoma kinase-positive T-cell lymphoma (ALK + TCL) cells from apoptosis. Proc. Natl. Acad. Sci. USA 2009, 106, 15843–15848. [Google Scholar]
  48. Brown, T.; Rushton, L. Occupational cancer in Britain. Haematopoietic malignancies: Leukaemia, multiple myeloma, non-Hodgkins lymphoma. Br. J. Cancer 2012, 107 Suppl 1, S41–S48. [Google Scholar]
  49. Zhao, X.F.; Reitz, M.; Chen, Q.C.; Stass, S. Pathogenesis of early leukemia and lymphoma. Cancer Biomark 2010, 9, 341–374. [Google Scholar]
  50. Carroll, M.C.; Katzman, P.; Alicot, E.M.; Koller, B.H.; Geraghty, D.E.; Orr, H.T.; Strominger, J.L.; Spies, T. Linkage map of the human major histocompatibility complex including the tumor necrosis factor genes. Proc. Natl. Acad. Sci. USA 1987, 84, 8535–8539. [Google Scholar]
  51. Harris, N.L.; Jaffe, E.S.; Diebold, J.; Flandrin, G.; Muller-Hermelink, H.K.; Vardiman, J. Lymphoma classification—from controversy to consensus: The R.E.A.L. and WHO Classification of lymphoid neoplasms. Ann. Oncol 2000, 11 Suppl 1, 3–10. [Google Scholar]
  52. Begg, C.B.; Mazumdar, M. Operating characteristics of a rank correlation test for publication bias. Biometrics 1994, 50, 1088–1101. [Google Scholar]
  53. Egger, M.; Davey Smith, G.; Schneider, M.; Minder, C. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997, 315, 629–634. [Google Scholar]
  54. Lau, J.; Antman, E.M.; Jimenez-Silva, J.; Kupelnick, B.; Mosteller, F.; Chalmers, T.C. Cumulative meta-analysis of therapeutic trials for myocardial infarction. N. Engl. J. Med 1992, 327, 248–254. [Google Scholar]
Figure 1. Overall association between TNF-α-308G>A polymorphism and NHL risk (additive model) in Caucasian and Asian populations. For each study, the estimate of odds ratio (OR) and its 95% confidence interval (CI) is plotted with a box and a horizontal line. The symbol diamond indicates pooled OR and its 95% CI.
Figure 1. Overall association between TNF-α-308G>A polymorphism and NHL risk (additive model) in Caucasian and Asian populations. For each study, the estimate of odds ratio (OR) and its 95% confidence interval (CI) is plotted with a box and a horizontal line. The symbol diamond indicates pooled OR and its 95% CI.
Ijms 15 07684f1
Figure 2. Begg’s funnel plot for publication bias on the association between TNF-α-308G>A polymorphism and NHL risk in additive model.
Figure 2. Begg’s funnel plot for publication bias on the association between TNF-α-308G>A polymorphism and NHL risk in additive model.
Ijms 15 07684f2
Table 1. Characteristics of 18 eligible articles included in this meta-analysis.
Table 1. Characteristics of 18 eligible articles included in this meta-analysis.
StudyEthnicityGenotyping MethodSamplesCharacteristics

NHLs (n)Controls (n)
Chouchane, 1997CaucasiansASPCR44106All subtypes
Demeter, 1997CaucasiansPCR-RFLP63117HCL only
Warzocha, 1998CaucasiansASPCR27396All subtypes
Fitzgibbon, 1999CaucasiansPCR-RFLP12188FL only
Wihlborg, 1999CaucasiansPCR-SPM4951CLL only
Mainou-Fowler, 2000CaucasiansPCR-RFLP7640CLL
Juszczynski, 2002CaucasiansSequencing204120All subtypes
Hellmig, 2005CaucasiansTaqMan138533MALT only
Bel Hadj Jrad, 2007CaucasiansPCR-RFLP194160All subtypes
Jevtovic-Stoimenov, 2008CaucasiansPCR-RFLP8034All subtypes
Fernberg, 2010CaucasiansSequenom22671484All subtypes
Skibola, 2010Caucasians, AsiansTaqMan or Pyrosequencing42875591All subtypes
Xiao, 2011AsiansPCR-RFLP160214All subtypes
Ibrahim, 2012CaucasiansPCR-RFLP84100BCL only
Hosgood, 2013AsiansTaqMan291300All subtypes
Lech-Maranda, 2013CaucasiansTaqMan288192CLL only
Liu, 2013AsiansPCR-LDR19323622TCL only
Nasira, 2013AsiansPCR-RFLP68129All subtypes
Abbreviations: NHL, non-Hodgkin lymphomas; ASPCR, allelic specific polymerase chain reaction; PCR-RFLP, polymerase chain reaction-restriction fragment length polymorphism; PCR-SPM, polymerase chain reaction-solid-phase minisequencing; PCR-LDR, polymerase chain reaction-ligation detection reaction; HCL, hairy cell leukemias; FL, follicular lymphomas; CLL, chronic lymphocytic leukemias; MALT, mucosal-associated lymphomas; BCL, B-cell lymphomas; TCL, T-cell lymphomas.
Table 2. Stratified analyses of TNF-α-308G/A polymorphism on NHL risk in Caucasians and Asians *.
Table 2. Stratified analyses of TNF-α-308G/A polymorphism on NHL risk in Caucasians and Asians *.
TypeEthnicityStudy (n)SamplesOR (95% CI)pI2 (%)phet

Cases (n)Controls (n)
NHLCaucasians14789384471.22 (1.06–1.40)0.00760.70.002
Asians5272645300.75 (0.66–0.86)<0.0010.00.670
Overall1910,61912,9771.06 (0.92–1.23)0.41375.0<0.001

BCLCaucasians11636980851.18 (1.03–1.34)0.01444.60.054
Asians2172338870.70 (0.52–0.94)0.01841.90.189
Overall13809211,9721.07 (0.91–1.26)0.41174.4<0.001

TCLCaucasians246768101.20 (1.01–1.42)0.0400.00.361
Asians263339220.96 (0.74–1.23)0.72357.80.124
Overall4110010,7321.11 (0.96–1.28)0.14543.10.153

DLBCLCaucasians3232569301.21 (1.11–1.32)<0.0010.00.491
Asians2102838870.70 (0.57–0.86)0.0010.00.908
Overall5335310,8170.97 (0.75–1.26)0.84083.9<0.001

FLCaucasians3123368981.00 (0.89–1.13)0.94931.00.235
Asians218438870.72 (0.47–1.12)0.14212.40.285
Overall5141710,7850.98 (0.87–1.10)0.70632.30.206

CLL/SLLCaucasians6185971271.02 (0.92–1.13)0.76713.80.326

MCLCaucasians225068101.25 (1.00–1.57)0.0520.00.560

MALTCaucasians11385331.07 (0.76–1.51)0.689

PTCLCaucasians118353261.11 (0.85–1.47)0.446
Asians1793000.65 (0.29–1.48)0.303
Overall226256361.05 (0.80–1.36)0.74132.90.222

NK/TCLAsians219039220.74 (0.46–1.17)0.19612.40.285
*Fixed-effect model was used when p value for heterogeneity test >0.05; otherwise, random-effect model was used;
Abbreviations: TNF, tumor necrosis factor; NHL, non-Hodgkin lymphomas; BCL, B-cell lymphomas; TCL, T-cell lymphomas; DLBCL, diffuse large B-cell lymphomas; FL, follicular lymphomas; CLL/SLL, chronic lymphocytic leukemias/small lymphocytic lymphomas; MCL, mantel cell lymphomas; MALT, mucosal-associated lymphomas; PTCL, peripheral T-cell lymphomas; NK/TCL, natural killer/T-cell lymphomas.
Table 3. Summary of primary data from eligible studies in this meta-analysis.
Table 3. Summary of primary data from eligible studies in this meta-analysis.
TypeEthnicityStudyCasesControls


GGGAAAGGGAAA
NHLCaucasiansChouchane, 19971133072331
Demeter, 19974218381342
Warzocha, 199820365569243
Fitzgibbon, 19999623264222
Wihlborg, 19992919137140
Mainou-Fowler, 20005023328111
Juszczynski, 200215149485323
Hellmig, 20059339636016013
Bel Hadj Jrad, 20071205915107494
Jevtovic-Stoimenov, 20083246219141
Fernberg, 20101490675102100743146
Skibola, 20102712113616437911394141
Ibrahim, 201241212267276
Lech-Maranda, 2013213678136533

AsiansSkibola, 2010243293212494
Xiao, 2011138202174355
Hosgood, 201317022219309150625
Liu, 2013264270260400
Nasira, 20135891105222

BCLCaucasiansDemeter, 19974218381342
Fitzgibbon, 19999623264222
Wihlborg, 19992919137140
Mainou-Fowler, 20005023328111
Juszczynski, 20027229385323
Hellmig, 20059339636016013
Jevtovic-Stoimenov, 20082429219141
Fernberg, 2010139563091100743146
Skibola, 2010222191513937911394141
Ibrahim, 201241212267276
Lech-Maranda, 2013213678136533

BCLAsiansSkibola, 2010194232212494
Hosgood, 201313321648309150625

TCLCaucasiansFernberg, 2010954511100743146
Skibola, 2010216901037911394141

AsiansHosgood, 2013287541309150625
Liu, 2013264270260400

DLBCLCaucasiansFernberg, 201037117323100743146
Juszczynski, 20027229385323
Skibola, 201010934956637911394141

AsiansSkibola, 201086112212494
Hosgood, 2013829973309150625

FLCaucasiansFitzgibbon, 19999623264222
Fernberg, 201029711512100743146
Skibola, 20104891673237911394141

AsiansSkibola, 20104860212494
Hosgood, 2013115132309150625

CLL/SLLCaucasiansWihlborg, 19992919137140
Mainou-Fowler, 20005023328111
Jevtovic-Stoimenov, 20082429219141
Fernberg, 201037317124100743146
Skibola, 20106051932537911394141
Lech-Maranda, 2013213678136533

MCLCaucasiansFernberg, 2010763310100743146
Skibola, 20109035637911394141

MALTCaucasiansHellming, 20059339636016013

PTCLCaucasiansSkibola, 201012553537911394141

AsiansLiu, 20137270260400

NK/TCLAsiansHosgood, 201389140309150625
Liu, 20138160260400
Abbreviations: NHL, non-Hodgkin lymphomas; BCL, B-cell lymphomas; TCL, T-cell lymphomas; DLBCL, diffuse large B-cell lymphomas; FL, follicular lymphomas; CLL/SLL, chronic lymphocytic leukemias/small lymphocytic lymphomas; MCL, mantle cell lymphomas; MALT, mucosal-associated lymphomas; PTCL, peripheral T-cell lymphomas; NK/TCL, NK/T-cell lymphomas.

Share and Cite

MDPI and ACS Style

Zhai, K.; Ding, J.; Zhou, Y. Different Role of Tumor Necrosis Factor-α Polymorphism in Non-Hodgkin Lymphomas among Caucasian and Asian Populations: A Meta-Analysis. Int. J. Mol. Sci. 2014, 15, 7684-7698. https://doi.org/10.3390/ijms15057684

AMA Style

Zhai K, Ding J, Zhou Y. Different Role of Tumor Necrosis Factor-α Polymorphism in Non-Hodgkin Lymphomas among Caucasian and Asian Populations: A Meta-Analysis. International Journal of Molecular Sciences. 2014; 15(5):7684-7698. https://doi.org/10.3390/ijms15057684

Chicago/Turabian Style

Zhai, Kan, Jie Ding, and Yan Zhou. 2014. "Different Role of Tumor Necrosis Factor-α Polymorphism in Non-Hodgkin Lymphomas among Caucasian and Asian Populations: A Meta-Analysis" International Journal of Molecular Sciences 15, no. 5: 7684-7698. https://doi.org/10.3390/ijms15057684

Article Metrics

Back to TopTop