GET THE APP

Single Nucleotide Variants and Somatic Aberrations of A20 in Immu
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Short Communication - (2016) Volume 7, Issue 5

Single Nucleotide Variants and Somatic Aberrations of A20 in Immune-Related Diseases and Lymphoid Neoplasms

Karoline Fechter1,2, Peter Neumeister1 and Alexander J A Deutsch1*
1Division of Hematology, Department of Internal Medicine, Medical University of Graz, Austria
2CBmed, Graz, Austria
*Corresponding Author: Alexander J A Deutsch, Stiftingtalstrasse 24, 8010 Graz, Austria, Tel: +43 316 385 72816, Fax: +43 316 385 73009 Email:

Abstract

A20- also called tumor necrosis factor (TNF) α induced protein 3 (TNFAIP3) - is an ubiquitin-modifying enzyme, which acts as negative regulator of the NF-κB pathway. Aberrant activation of NF-κB has been shown to contribute to chronic inflammation and development of cancer. Somatic mutations and epigenetic silencing at A20 have been recently identified to be involved in the development of classical Hodgkin and many other types of non- Hodgkin lymphomas, thereby rendering A20 a classical tumor suppressor. Moreover, single nucleotide polymorphisms of A20 have been described to participate in the development of autoimmune diseases of the intestinal tract, systemic lupus erythematosus, rheumatoid arthritis and diabetes mellitus Type I. Here we want to summarize different polymorphisms related to autoimmune disorders and mention other SNPs, mutations and epigenetic mechanisms affecting A20 in lymphomas and multiple myeloma.

Keywords: A20; NF-kB; Single nucleotide polymorphisms; Somatic aberrations; Epigenetic silencing; Autoimmune disease; Lymphoid neoplasms

A20 Structure

A20, also known as tumor necrosis factor (TNF) a induced protein 3 (TNFAIP3), is an ubiquitin-modifying enzyme, which acts as negative regulator of NF-κB. The protein was first identified in 1990 as a cytokine response gene in human umbilical vein endothelial cells [1]. A20 is located on chromosome 6 and has eight coding exons [2]. As shown in Figure 1 the C-terminal region contains seven zinc finger (ZnF) domains, whereby domain 4 harbors an ubiquitin-binding and E3 ligase activity and ZnF 7 has an additional ubiquitin-binding activity. The N-terminal region is characterized by an ovarian tumor domain (OTU) responsible for deubiquitinylation. The deubiquitinating (DUB) activity is thereby mediated by a catalytic cysteine residue at position C103 of A20 [3,4]. Importantly, A20 induction can be mediated by various nuclear factor kappa-lightchain- enhancer of activated B cells (NF-κB)-mediated stimuli through NF-κB binding sites in the A20 promotor region and functions itself as a negative regulator of the same pathway thereby mediating a regulatory crosstalk [5].

clinical-cellular-immunology-protein-structure

Figure 1: Schematic representation of the A20 protein structure - adapted from Ma et al. [4] At the N-terminal site the A20 protein has an ovarian tumor domain (OTU) harbouring a deubiquitinating (DUB) activity, which is mediated by a cysteine residue at position C103. At the C-terminal domain the protein contains seven zinc finger (ZnF) regions with an E3 ubiquitin ligase activity on zinc finger 4 and ubiquitinbinding regions lying on ZnF 4 and ZnF 7, respectively.

A20 as a negative regulator of the NF-κB pathway

A20 is mainly regulated by the CARMA1-BCL10-MALT1 complex, which connects T-cell receptor signalling with the canonical NF-κB pathway [3]. The way by which A20 inhibits NF-κB signaling is very complex and unique. Like other E3 ligases and deubiquitinating enzymes, A20 is involved in ubiquitinylation of several proteins [6]. The polyubiquitinylation process represents a posttranscriptional modification, which causes proteasomal degradation. The OTU domain of A20 cleaves lysine-63 linked polyubiquitin chains from the receptor-interaction protein 1 (RIP1), which is an essential factor for the tumor necrosis factor (TNF) receptor 1 (TNFR1) signaling pathway. This reaction is followed by adding lysin-43 linked polyubiquitin chains and subsequent degradation of RIP1 through the E3 ligase activity of A20 [5]. This suggests that A20 has a double function in the TNFR1 pathway [7]. Furthermore, A20 does not only regulate the ubiquitinylation of RIP1 in the TNFR1 pathway but it also suppresses the activation of NF-κB through TNF by deubiquitinating TRAF6 [8,9].

A20 and autoimmune diseases

One of the most important and best-characterized pathways involved in innate and adaptive immunity and tumorigenesis is the canonical NF-κB pathway, whose constitutive activation leads to proinflammatory processes. A20 is induced by NF-κB itself and functions upon induction as an auto-negative feedback regulator of the pathway. With respect to its important regulatory role constricting overwhelming immune responses, it has been identified as a susceptibility gene for multiple inflammatory processes as well as autoimmune diseases. It is assumed that single-nucleotide polymorphisms (SNPs) of A20 favor pro-inflammatory processes through its reduced general anti-inflammatory potential. The SNPs affecting A20 and being involved in autoimmune diseases are distributed throughout the whole protein sequence, thereby affecting the N- and C-terminal domains as well as coding and non-coding regions of the latter, leading to reduced function and/or expression of A20. Several genome-wide association studies (GWAS) and other smaller genomic studies identified several distinct polymorphisms of A20 located on the human chromosomal region 6q.23. Generally, polymorphisms of A20 seem to be important in intestinal homeostasis and mucosal biology of the gut. NF-κB and thus A20 are key players in mucosal integrity participating in control of inflammatory processes and inflammatory cytokines secreted by resident immune cells. Evidence for a role of A20 in intestinal biology comes from genomic studies of different populations, where polymorphisms of A20 have been shown to be implicated in inflammatory bowel disease (IBD), Crohn’s disease and coeliac disease. Genomic analysis of Caucasian and British population showed a reduced or defective function of A20 in IBD and Crohn’s disease [4,10-16]. Furthermore, SNPs in the A20 locus have been identified to be predisposing for the development of other autoimmune disorders like rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). In RA excessive secretion of proinflammatory TNFa plays a key role in disease progression and TNFa antagonists serve as therapeutic targets for the treatment of this autoimmune disorder. SLE patients represent reduced self-tolerance and increased interferon levels. Both diseases share -at least partiallypolymorphisms affecting the same nucleotides. Two SNPs, rs2230926/ F127C and rs5029941/ A125V, both lying in exon 3 in the OTU domain, have been shown to be important in SLE. A125V has been associated with increased risk of IBD in African-Americans, whereas it seems to have a protective function in SLE [17], while F127C has been identified to negatively affect the inhibitory capacity of A20 in SLE. Moreover, the SNP F127C has been meanwhile detected in a variety of different autoimmune diseases like psoriasis, Crohn’s disease, Sjörgen’s syndrome and RA, respectively, whereas the SNP rs6920220 has been shown to be a risk allele for RA and SLE only [18-22].

Type 1 diabetes (T1D) represents another autoimmune disease, where death of pancreatic ß-islets is caused by the accumulation of inflammatory mediators in whose expression NF-κB and A20 as negative regulator of NF-κB are involved. A study of Fung et al. showed, that polymorphisms identified also in RA, SLE and Crohn’s disease, could be also detected in T1D [23]. A list of different SNPs affecting A20 expression and/ or function in immune-related processes is depicted in Table 1. Noteworthy, several SNPs seem to be implicated in more than one autoimmune disease, while others might have protective or negative effects depending on disease type and population [21].

SNP Location Nucleotide Position Disease association Population
rs13207033 223 kb upstream A/ G 137965418 RA American
rs13192841 221 kb upstream A/ G 137967214 SLE European
Sjörgen’ssyndrom European
rs2327832 215 kb upstream A/ G 137973068 Coeliac Disease European
RA European
rs10499194 186 kb upstream C/ T 138002637 RA European/ African-American, American, European
Juvenile idiopathic arthritis Japanese
SLE/ RA European
Type 1 diabetes European
rs6920220 182 kb upstream A/ G 138006503 RA American,European,European/ African-American
Juvenile idiopathic arthritis European
Type 1 Diabetes European
SLE European
rs7753394 103 kb upstream C/ T 138085248 Crohn’s disease European
rs10499197 56 kb upstream G/ T 138132516 SLE European
rs5029930 Intron 1 A/ C 138190684 Coronary artery disease in Type 2 diabetes American
rs5029937 Intron 2 G/ T 138195151 RA European
rs5029939 Intron 2 C/ G 138195723 Systemic sclerosis European
SLE European/ African-American
rs5029941 Exon 3 C/ T (A125V) 138196060 SLE/ IBD African-American
rs2230926 Exon 3 T/ G (F127C) 138196066 SLE European, African-American,Japanese,Chinese
SLE/ RA Japanese
Sjörgen’s syndrome/ Crohn’s disease/ psoriasis/ RA European
rs582757 Intron 5 A/ G 138197824 Rheumatic heart disease Chinese
New (no rs) Intron 5 C/ - 138197889 RA European
rs610604 Intron 6 A/ C 138199417 Psoriasis Caucasian
Coronary artery diseases in Type 2 diabetes American
rs5029953 Intron 7 A/ G 138200760 SLE African-American
rs7749323 26 kb downstream A/ G 138230389 SLE European
rs5029930 28 kb downstream A/ C 138232377 Coronary artery disease in Type 2 diabetes American
Polymorphic dinucleotide 68 kb downstream TT>A 138272732-138271733 SLE European/ Korean
rs6922466 256 kb downstream A/ G 138444930 SLE European
Sjörgen’ssyndrom European

Table 1: Genetic variants of A20 related to autoimmune disease as adapted from Vereecke et al. and Nocturne et al. [11,12,22].

A20 and hematological neoplasms

A20 is known to act as a tumor suppressor in lymphoid malignancies. Aberrant NF-κB activation is implicated in tumor development and progression and recent studies have proven evidence that somatic mutations or epigenetic mechanisms affecting A20 are involved in the development and progression of several lymphoid malignancies. Somatic mutations and/ or deletions as well as promotor methylation of A20 lead to its inactivation and consequently to an over-activated NF-κB pathway in malignant cells with a reduced apoptotic potential due to an enhanced expression of anti-apoptotic genes. Importantly, accumulation of proliferating cells combined with the reduced ability to eliminate them, favors the augmentation of mutations within a given proliferating cell population and thus gives rise to further malignant somatic transformations. The mutations are not clustered into a specific region of the protein, rather they affect different regions ranging from the C-terminal OTU domain to the Nterminal Zn-Finger domains [10]. Likewise, the mutations observed can be varying, ranging from deletions, nonsense mutations introducing premature stop codons, missense mutations, frameshift mutations or insertions to nucleotide substitutions at splice donor sites. Several articles published within the last decade have highlighted the involvement of A20 in non-Hodgkin and classical Hodgkin lymphomas as well as Burkitt’s lymphoma, natural killer (NK) cell lymphoma, and adult T cell leukaemia (ATLL). Mono- and biallelic inactivation by deletions at 6q23.3 as well as different somatic mutations could be detected in mucosa-associated lymphoid tissue (MALT) lymphomas of the ocular adnexa, salivary gland and thyroidbut not of the lung, stomach or skin- , mantle cell lymphoma (MZL), activated B cell or germinal centre B cell like diffuse large B cell lymphomas (ABC-DLBCL and GCB-DLBCL, respectively), primary mediastinal B cell lymphomas (PMBL), marginal zone lymphomas (MZL), follicular lymphomas and classical Hodgkin lymphomas (cHL) [24-33]. Interestingly, the occurrence of monoallelic deletions, having an inhibiting effect on A20 expression and function, indicates that A20 deletion might also act in a haplo-insufficient way and/ or in combination with gene amplification due to mutations in other proteins positively influencing NF-κB signalling like caspase recruitment domain-containing protein 11 (CARD11) [34]. Moreover, changes in A20 function have been attributed to epigenetic mechanisms like promotor methylation, which were particularly found in MCL, MALT lymphomas and lymphomas of the ABC- and GCBDBCL subtype [26,29].

Recent work by our group could show that monoallelic deletions together with an increased expression of NF-κB target genes could be found in multiple myeloma (MM), another type of haematological malignancy [35]. Additionally we were able to detect a germline SNP rs143002189 in MM, which leads to a missense single base pair substitution - c.2364G>A- and predisposes to de development of DLBCL [36]. The latter could also be found in HL and benign hemangioblastomas [30,37].

Concluding Remarks

A20 is known to act as a tumor suppressor in many pathogenic processes ranging from auto-destructive and still difficult-to-treat autoimmune diseases to severe and prone-to-relapse haematological neoplasms. Several single nucleotide polymorphisms affecting A20 could be identified as disease-related in autoimmune-diseases like RA, SLE and malignancies affecting the homeostasis of the gut microflora. Both, somatic mutations and deletions as well as epigenetic mechanisms seem to be responsible for inactivation of A20 in haematological neoplasms of non-Hodgkin and Hodgkin lymphomas. Nevertheless, the role of many germline single nucleotide polymorphisms still remain to be elucidated in haematological malignancies, a fact, which would certainly impact risk stratification and treatment options.

Funding

KF was supported by CBmed. PN was supported by grants of Fellinger Krebsforschung, Land Steiermark, Jubiläumsfond der ÖNB (N11181). AD was supported by the START-Funding-Program of the Medical University of Graz and by a research grant of MEFOgraz.

References

  1. Dixit VM, Green S, Sarma V, Holzman LB, Wolf FW, et al. (1990) Tumor necrosis factor-alpha induction of novel gene products in human endothelial cells including a macrophage-specific chemotaxin. J BiolChem 265: 2973-2978.
  2. Verstrepen L, Verhelst K, van Loo G, Carpentier I, Ley SC, et al. (2010) Expression, biological activities and mechanisms of action of A20 (TNFAIP3). BiochemPharmacol 80: 2009-2020.
  3. Zhang F, Yang L, Li Y (2012) The role of A20 in the pathogenesis of lymphocytic malignancy. Cancer Cell Int 12: 44.
  4. Ma A, Malynn BA (2012) A20: linking a complex regulator of ubiquitylation to immunity and human disease. Nat Rev Immunol 12: 774-785.
  5. Shembade N, Harhaj E (2012) Regulation of NF-kappaB signaling by the A20 deubiquitinase. Cell MolImmunol 9: 123 -130.
  6. Malynn BA, Ma A (2009) A20 takes on tumors: tumor suppression by an ubiquitin-editing enzyme. J Exp Med 206: 977-980.
  7. Wertz IE, O'Rourke KM, Zhou H, Eby M, Aravind L, et al. (2004) De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaBsignalling. Nature 430: 694-699.
  8. Boone DL, Turer EE, Lee EG, Ahmad RC, Wheeler MT, et al. (2004) The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol 5: 1052-1060.
  9. Turer EE, Tavares RM, Mortier E, Hitotsumatsu O, Advincula R, et al. (2008) Homeostatic MyD88-dependent signals cause lethal inflamMation in the absence of A20. J Exp Med 205: 451-464.
  10. Hymowitz SG, Wertz IE (2010) A20: from ubiquitin editing to tumour suppression. Nat Rev Cancer 10: 332-341.
  11. Vereecke L, Beyaert R, van Loo G (2011) Genetic relationships between A20/TNFAIP3, chronic inflammation and autoimmune disease. BiochemSoc Trans 39: 1086-1091.
  12. Vereecke L, Beyaert R, van Loo G (2009) The ubiquitin-editing enzyme A20 (TNFAIP3) is a central regulator of immunopathology. Trends Immunol 30: 383-391.
  13. Wellcome Trust Case Control Consortium (2007) Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447: 661-678.
  14. Arsenescu R, Bruno ME, Rogier EW, Stefka AT, McMahan AE, et al. (2008) Signature biomarkers in Crohn's disease: toward a molecular classification. Mucosal Immunol 1: 399-411.
  15. Trynka G, Zhernakova A, Romanos J, Franke L, Hunt KA, et al. (2009) Coeliac disease-associated risk variants in TNFAIP3 and REL implicate altered NF-kappaBsignalling. Gut 58: 1078-1083.
  16. Barmada MM, Brant SR, Nicolae DL, Achkar JP, Panhuysen CI, et al. (2004) A genome scan in 260 inflammatory bowel disease-affected relative pairs. Inflamm Bowel Dis 10: 513-520.
  17. Lodolce JP, Kolodziej LE, Rhee L, Kariuki SN, Franek BS, et al. (2010) African-derived genetic polymorphisms in TNFAIP3 mediate risk for autoimmunity. J Immunol 184: 7001-7009.
  18. Orozco G, Hinks A, Eyre S, Ke X, Gibbons LJ, Bowes J, et al. (2009) Combined effects of three independent SNPs greatly increase the risk estimate for RA at 6q23. Hum Mol Genet 18: 2693-2699.
  19. Graham RR, Cotsapas C, Davies L, Hackett R, Lessard CJ, et al. (2008) Genetic variants near TNFAIP3 on 6q23 are associated with systemic lupus erythematosus. Nat Genet 40: 1059-1061.
  20. Musone SL, Taylor KE, Lu TT, Nititham J, Ferreira RC, et al. (2008) Multiple polymorphisms in the TNFAIP3 region are independently associated with systemic lupus erythematosus. Nat Genet 40: 1062-1064.
  21. Musone SL, Taylor KE, Nititham J, Chu C, Poon A, et al. (2011) Sequencing of TNFAIP3 and association of variants with multiple autoimmune diseases. Genes Immun 12: 176-182.
  22. Nocturne G, Boudaoud S, Miceli-Richard C, Viengchareun S, Lazure T, et al. (2013) Germline and somatic genetic variations of TNFAIP3 in lymphoma complicating primary Sjogren's syndrome. Blood 122: 4068-4076.
  23. Fung EY, Smyth DJ, Howson JM, Cooper JD, Walker NM, et al. (2009) Analysis of 17 autoimmune disease-associated variants in type 1 diabetes identifies 6q23/TNFAIP3 as a susceptibility locus. Genes Immun 10: 188-191.
  24. Chanudet E, Ye H, Ferry J, Bacon CM, Adam P, et al. (2009) A20 deletion is associated with copy number gain at the TNFA/B/C locus and occurs preferentially in translocation-negative MALT lymphoma of the ocular adnexa and salivary glands. J Pathol 217: 420-430.
  25. Chanudet E, Huang Y, Ichimura K, Dong G, Hamoudi RA, et al. (2010) A20 is targeted by promoter methylation, deletion and inactivating mutation in MALT lymphoma. Leukemia 24: 483-487.
  26. Honma K, Tsuzuki S, Nakagawa M, Tagawa H, Nakamura S, et al. (2009) TNFAIP3/A20 functions as a novel tumor suppressor gene in several subtypes of non-Hodgkin lymphomas. Blood 114: 2467-2475.
  27. Kato M, Sanada M, Kato I, Sato Y, Takita J, et al. (2009) Frequent inactivation of A20 in B-cell lymphomas. Nature 459: 712-716.
  28. Compagno M, Lim WK, Grunn A, Nandula SV, Brahmachary M, et al. (2009) Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature 459: 717-721.
  29. Paik JH, Go H, Nam SJ, Kim TM, Heo DS, et al. (2013) Clinicopathologic implication of A20/TNFAIP3 deletion in diffuse large B-cell lymphoma: an analysis according to immunohistochemical subgroups and rituximab treatment. Leuk Lymphoma 54: 1934-1941.
  30. Schmitz R, Hansmann ML, Bohle V, Martin-Subero JI, Hartmann S, et al. (2009) TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. J Exp Med 206: 981-989.
  31. Schumacher MA, Schmitz R, Brune V, Tiacci E, Doring C, et al. (2010) Mutations in the genes coding for the NF-kappaB regulating factors IkappaBalpha and A20 are uncommon in nodular lymphocyte-predominant Hodgkin's lymphoma. Haematologica 95: 153-157.
  32. Novak U, Rinaldi A, Kwee I, Nandula SV, Rancoita PM, et al. (2009) The NF-{kappa}B negative regulator TNFAIP3 (A20) is inactivated by somatic mutations and genomic deletions in marginal zone lymphomas. Blood 113: 4918-4921.
  33. Thelander EF, Ichimura K, Corcoran M, Barbany G, Nordgren A, et al. (2008) Characterization of 6q deletions in mature B cell lymphomas and childhood acute lymphoblastic leukemia. Leuk Lymphoma 49: 477-487.
  34. Bu R, Bavi P, Abubaker J, Jehan Z, Al-Haqawi W, et al. (2012) Role of nuclear factor-κB regulators TNFAIP3 and CARD11 in Middle Eastern diffuse large B-cell lymphoma. Leuk Lymphoma 53: 1971-1977.
  35. Troppan K, Hofer S, Wenzl K, Lassnig M, Pursche B, et al. (2015) Frequent down regulation of the tumor suppressor gene a20 in multiple myeloma. PLoS One 10: e0123922.
  36. Wenzl K, Hofer S, Troppan K, Lassnig M, Steinbauer E, et al. (2016) Higher incidence of the SNP Met 788 Ile in the coding region of A20 in diffuse large B cell lymphomas. TumourBiol 37: 4785-4759.
  37. Shankar GM, Taylor-Weiner A, Lelic N, Jones RT, Kim JC, et al. (2014) Sporadic hemangioblastomas are characterized by cryptic VHL inactivation. ActaNeuropatholCommun 2: 167.
Citation: Fechter K, Neumeister P, Deutsch AJA (2016) Single Nucleotide Variants and Somatic Aberrations of A20 in Immune-Related Diseases and Lymphoid Neoplasms. J Clin Cell Immunol 7:452.

Copyright: © 2016 Fechter K, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top