1,374
Views
14
CrossRef citations to date
0
Altmetric
Extra Views

APOBEC3 proteins and genomic stability

The high cost of a good defense

, &
Pages 33-38 | Received 27 Oct 2011, Accepted 08 Nov 2011, Published online: 01 Jan 2012

Abstract

The human APOBEC3 family of cytidine deaminases constitutes a cellular intrinsic defense mechanism that is effective against a range of viruses and retro-elements. While it is well established that these enzymes are powerful mutators of viral DNA, the possibility that their activity could threaten the integrity of the host genome has only recently begun to be investigated. Here, we discuss the implications of new evidence suggesting that APOBEC3 proteins can mediate the deamination of cellular DNA. The maintenance of genomic integrity in the face of this potential off-target activity must require high fidelity DNA repair and strict regulation of APOBEC3 gene expression and enzyme activity. Conversely, the ability of specific members of the APOBEC3 family to activate DNA damage signaling pathways might also reflect another way that these proteins contribute to the host immune response.

APOBEC3 Proteins in Innate Immunity

The AID/APOBEC3 family of cytidine deaminases has emerged as an important component of the immune system in vertebrates, acting in both innate and adaptive pathways.Citation1 The AID/APOBEC3 enzymes share the ability to catalyze the deamination of cytidine to uracil and are thus capable of functioning as powerful DNA mutators.Citation2 These cytidine deaminases have evolved to target the genome of both the host and its invading pathogens. Activation-induced cytidine deaminase (AID), a key mediator of the adaptive humoral immune response, functions in germinal center B cells to diversify antigen receptor genes through the processes of somatic hypermutation (SHM) and class switch recombination (CSR).Citation3,Citation4 In contrast, the human apolipoprotein-B mRNA-editing catalytic polypeptide-like 3 (APOBEC3) family of cytidine deaminases, which comprise seven host restriction factors (APOBEC3A-H), constitute an innate barrier to retroviruses, endogenous retro-elements and DNA viruses.Citation5

APOBEC3 proteins have been shown to exert selective antiviral activity against a range of viruses and to act at specific steps within the viral infection cycle.Citation6 Several studies have demonstrated that the restricting action of APOBEC3 proteins can result from both deaminase-dependent and deaminase-independent mechanisms.Citation7 Nevertheless, in many cases, APOBEC3-mediated antiviral activity correlates with the detection of hypermutated viral genomes.Citation8Citation13 Hypermutation of viral genomes by APOBEC3 proteins has been suggested to compromise viral infectivity by altering the integrity of open reading frames. Alternatively, uracilcontaining viral DNA could represent a substrate for the cellular DNA cellular repair machinery. Deamination of cytosines creates uracil lesions that can be processed by the cellular DNA glycosylases, which catalyze the hydrolysis of the glycosylic bond between the base and the deoxyribose backbone, leaving an abasic site. It has been suggested that the concerted action of uracil-DNA glycosylase (UNG), the main glycosylase initiating the base-excision repair of uracil in DNA, and apurinic/apyrimidinic endonuclease1 (APE1), which catalyzes strand cleavage at the lesion, can trigger the degradation of deaminated viral DNA.Citation8,Citation14 However, this hypothesis has been challenged,Citation15Citation17 and it remains unclear whether these repair enzymes contribute to APOBEC3-mediated antiviral activity.

In addition to their role as antiviral factors, it has been recently proposed that APOBEC3 proteins can mediate the extensive deamination and degradation of transfected plasmid DNA.Citation18 A study by Stenglein et al.Citation18 described the ability of several APOBEC3 members to restrict gene transfer and suggested that clearance of foreign DNA could be a distinct physiological function of APOBEC3 proteins. Among all APOBEC3 proteins, APOBEC3A (A3A) had the strongest DNA-restricting ability,Citation18 consistent with its previously reported potent cytidine deaminase activity.Citation19 Using selective amplification of edited DNA through differential denaturation (3D)-PCR,Citation20 deamination marks were detected in plasmids recovered from transfected primary macrophages that were induced to express A3A. The authors were, however, unable to detect editing in genomic DNA, leading them to suggest that the host nuclear DNA may be less susceptible to A3A-mediated deamination than plasmid DNA.

Collateral Damage from Cytidine Deaminases

The AID and APOBEC3 proteins have been assigned to specific physiological functions through their activities on defined substrates, i.e., AID functions at the immunoglobulin loci to promote antibody gene diversification, while APOBEC3 proteins act upon viral nucleic acids as a form of intrinsic host defense.Citation21 Their ability to trigger nucleotide alterations confers intrinsic mutagenic potential, implying that deamination of DNA outside of their intended targets could cause collateral damage to the cellular genome with dire consequences.Citation22 The precise details of how AID is targeted to the immunoglobulin genes are still being deciphered, but it is now clear that AID deaminates many more genes than originally suspected and can cause off-target mutations throughout the genome.Citation23,Citation24 Recent work has demonstrated that AID can deaminate many oncogenes involved in B-cell tumorigenesis, highlighting the role of base excision repair (BER) and mismatch repair (MMR) pathways in preventing the accumulation of deleterious mutation.Citation25 AID can also generate double-strand breaks (DSBs) at many non-immunoglobulin genes.Citation26 Processing of these breaks can lead to rare translocations through joining of DSBs generated during antigen receptor diversification to breaks that are located near oncogenes.Citation27,Citation28 In this regard, it has been shown that homologous recombination repair proteins act as safeguards to prevent genomic instability by the widespread genomic breaks that can be induced by off-target activity of AID in B cells.Citation29 Interestingly, it has been hypothesized that over the course of evolution, AID activity has been restricted to minimize the risk of genomic stability.Citation30

Evidence for APOBEC3-Mediated Deamination of Cellular DNA

The studies of APOBEC3 proteins have so far predominantly focused on understanding their antiviral activities and biochemical properties. In contrast, little is known about the possible detrimental consequences of expressing these mutagenic enzymes and their propensity to act in an “off-target” fashion on host rather than viral genomes. In a recent study, we reported that A3A expression can induce DNA breaks and activate the cellular DNA damage response (DDR) in a deaminasedependent fashion.Citation31 The DDR induced by A3A was revealed using antibodies generated to specific phosphorylation marks on DNA damage/repair proteins (such as the histone variant H2AX),Citation32 and DNA breaks were detected using the terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) assay. In addition, using a UNG inhibitor (UGI), we demonstrated that UNG activity is required for both A3A-induced DNA breaks and activation of the DDR, suggesting that DNA damage signaling occurs as a consequence of cellular processing of A3A-induced uracil lesions. Importantly, using stable cell lines expressing A3A under the control of a Tet-inducible promoter, we showed that activation of the DDR by induction of A3A expression is independent of the presence of foreign DNA. This observation implies that uncontrolled A3A expression can damage the cellular DNA in the absence of foreign DNA or virus infection. In our study, we also reported that ectopic A3A expression leads to cell cycle arrest in S phase. Similar observations have been made when B cells deficient for homologous recombination (HR) were induced to express AID.Citation29 Although it is unclear whether endogenous A3A can affect cell cycle progression, our results suggest that the effects of A3A expression on the host cell could have been a contributing factor in previous experiments focused on understanding A3A function as a restriction factor.

The precise series of events that results in activation of the DDR we detected with overexpressed A3A remains to be determined. Since A3A and the other AID/APOBEC3s have been well-documented to have a preference for single-stranded DNA substrates,Citation19,Citation33Citation35 the initiating deamination must occur on genomic DNA that is single-stranded and accessible. Active transcription is a prerequisite for somatic hypermutation induced by AID,Citation36Citation39 and thus, it is possible that the genomic instability induced by A3A is also associated with transcription. In this case, DNA damage could result from deamination on the coding DNA strand that transiently adopts a single-stranded conformation during the passage of a transcription bubble.Citation40 Another possibility is that the DNA damage induced by A3A occurs in S phase, when single-strand DNA is exposed during cellular DNA replication. This conclusion is consistent with our recent findings showing that when we induce A3A expression in cells arrested in G1, we do not detect H2AX phosphorylation (unpublished observations). Alternatively, it is possible that deaminated residues induced in G1 are not processed into breaks or are somehow protected from activation of the DDR. Identification of the nature of the genomic DNA substrate targeted by A3A will provide information that could lead to a better understanding of its physiological function.

While our work demonstrated that ectopic A3A expression is detrimental to cellular DNA integrity, we did not obtain conclusive evidence for activation of DNA damage signaling by endogenous A3A. Using an approach based on 3D-PCR, Suspene et al.Citation41 reported that mitochondrial DNA containing signs of deamination in the form of G→A and C→T hypermutations can be amplified from peripheral blood mononuclear cells expressing A3 proteins. In addition, the authors detected hypermutated nuclear DNA in cells from patients with a specific form of hyper-IgM syndrome that is associated with UNG deficiency.Citation42 These intriguing results suggest that cellular DNA is sensitive to endogenous levels of APOBEC3 proteins. However, further studies are required to evaluate the extent of APOBEC3-mediated mutagenesis of cellular DNA under physiological conditions.

Regulation of APOBEC3 Activity

The finding that APOBEC3 proteins are capable of editing cellular DNA provides a compelling reason to suggest that their deaminase activity must be tightly regulated. Since the discovery of AID and its role in SHM and CSR,Citation43Citation45 an immense amount of effort has been directed toward deciphering how the enzyme functions and determining how it is regulated.Citation46 Regulation has been proposed at many different levels, including transcription,Citation47 mRNA degradation by miRNAs,Citation48 cellular localization and active nuclear shuttling,Citation49 post-translational modifications,Citation50,Citation51 proteasomal degradationCitation52 and by interaction with various cellular partners.Citation53Citation55 It is intriguing to note that AID expression is augmented in B cell-derived lymphomas and leukemias,Citation56 and there is increasing evidence that AID upregulation by proinflammatory cytokines in tissues other than B cells can promote tumorigenesis.Citation57

In the case of APOBEC3 proteins, little is currently known about their regulation. APOBEC3 proteins have been reported to be expressed in a range of hematopoietic cells and to be induced in response to different cytokines and hormones.Citation19,Citation58Citation63 Notably, APOBEC3 gene expression was shown to be induced by type-I interferon (IFN) stimulation, consistent with their roles in the antiviral immune response. A limited number of studies have investigated the transcriptional regulation of the APOBEC3 gene,Citation64Citation67 and it is still unclear which transcription factors regulate its expression in response to IFN. A3A gene expression is very sensitive to INFαCitation61 and contains an interferon-stimulated response element (ISRE) in its potential promoter region.Citation58 Characterization of the APOBEC3 gene promoter regions is required, since a better understanding of their transcriptional regulation might provide crucial information on how the expression of A3 proteins can be modulated to reach a precarious costbenefit balance.

While APOBEC3G (A3G) activity has been shown to be regulated by several mechanisms,Citation5,Citation62,Citation68Citation80 the mechanisms controlling the activity of APOBEC3 proteins that localize to the nucleus have yet to be described. Is it, indeed, surprising that despite being the most potent deaminase of the APOBEC3 family, A3A partially localizes to the nucleus.Citation19,Citation81 While it has been reported to co-purify with complexes containing LINE-1 RNA,Citation82 interacting proteins that might regulate A3A activity have yet to be identified. It is indeed possible that A3A DNA binding affinity and/or activity are modulated by its interaction with a cellular co-factor(s), as has been proposed for other cytidine deaminases.Citation55,Citation83Citation86 Human A3A is expressed as two different isoforms, the smaller form resulting from the presence of an internal translation initiation codon.Citation87,Citation88 In a recent study, Thielen et al.Citation88 showed that both A3A isoforms appear to be enzymatically active. However, it remains unclear if the expression of two A3A isoforms represents a strategy to regulate its activity and whether the acquisition of an alternative start codon is a consequence of recent evolution.Citation88,Citation89

The finding that A3A-induced mutations in nuclear DNA are detectable exclusively in UGI-expressing cells suggests that the BER machinery is normally able to limit the deleterious effects of A3A activity.Citation41,Citation90 According to a recent analysis of APOBEC3 gene expression in hematopoietic cells, A3A is the sole member of the family whose expression is restricted to the myeloid lineage.Citation91 These observations support the hypothesis that restricting A3A expression to non-proliferating cells could constitute a strategy to limit the negative consequences of its off-target activity. In this regard, it has been reported that the A3A gene is under negative selection,Citation92 supporting the idea that, at some point during vertebrate evolution, the collateral damage resulting from A3A expression might have exceeded its beneficial properties. Similar hypotheses have been proposed to explain the frequent deletion of the APOBEC3B gene in humansCitation93 and the recent acquisition of destabilizing mutations affecting the anti-retroelement activity of APOBEC3H.Citation94

Linking the DDR and the Immune Response

Our recent analysis of A3A-induced DDR revealed that activation of many DNA damage mediators is exquisitely sensitive to the presence of A3A and can be observed in response to protein levels that are comparable to physiological A3A levels. It has been proposed that activation of the DDR could participate in the immune response by inducing the expression of ligands for the activating receptor NKGD2.Citation95,Citation96 In addition, Gourzi et al.Citation97 have shown that expression of AID in response to the transforming retrovirus Abelson murine leukemia virus (Ab-MLV) can activate DNA damage signaling and promote the expression of the NKG2D ligand Rae-1. The authors also found that virus-induced AID expression restricts the proliferation of Ab-MLV-infected cells. More recently, A3G expression in HIV-infected T cells has been shown to correlate with the upregulation of natural killer (NK) cell-activating ligands and activation of DNA damage signaling.Citation98 Interestingly, this study reported that uracil residues can be generated in both viral and cellular DNA in infected cells, and that this process is inhibited in the presence the viral Vpr and Vif proteins. Although it is unclear how A3G can access nuclear DNA in HIV-infected cells, these observations suggest that activation of the DDR by APOBEC3 proteins can promote the recognition of infected cells by NK cells.

In light of these observations, it is tempting to speculate that the controlled ability of A3A to activate the cellular DDR could similarly represent a strategy that contributes to the immune response. AID expression in germinal center B cells can affect cell viability, and this appears to correlate with its ability to induce DNA breaks.Citation99 This effect has been proposed to play a role in maintaining the integrity of the germinal center compartment and preventing the development of autoimmune B cells. It is thus possible that induction of DNA damage by members of the APOBEC3 family could contribute to cell cycle checkpoint activation and promote apoptosis in physiological conditions, such as in virus-infected cells.Citation96,Citation100 We have observed that prolonged induction of A3A expression in an inducible cell line leads to cell death (unpublished observations). It will be important to determine if endogenous A3A retains its damaging ability in certain cell types and whether it proves to be beneficial under specific circumstances.

Conclusions

The finding that APOBEC3 proteins can deaminate cellular DNA suggests they may contribute to genomic instability. The new observations we have here discussed, combined with the established links between AID and genomic instability, highlight the need to carefully investigate the potential role of APOBEC3 proteins in tumorigenesis and cancer. Genome protection from deaminase-induced mutagenesis requires tight regulation of the activity of the APOBEC3 proteins. It will be critical to identify the cellular mechanisms regulating APOBEC3 protein activity and determine whether uncontrolled deaminase activity can contribute to malignancy.Citation101Citation103 Stimulation of A3A expression as part of the immune response to pathogens provides necessary intrinsic antiviral defenses but could also promote tumorigenic pathways. Several mechanisms could participate in protecting genomic DNA from destructive deamination, including chromatin structure, subnuclear compartmentalization or steric hindrance by single-stranded DNA binding proteins that cover the DNA substrate. Little is currently known about A3A regulation by either transcriptional control or post-translational modifications, and cellular factors that associate with the protein to influence its activity remain to be identified. In addition to their well-documented antiviral activities, APOBEC3 proteins may provide some additional physiological function that requires expression in particular cell types. This might be restriction of foreign DNACitation18 or a recently suggested role in DNA demethylation.Citation104,Citation105 Although more work is needed to clearly establish the role of APOBEC3 proteins in these processes, it is also intriguing to consider that deamination of the host cell DNA could be advantageous, and that APOBEC3 function in the immune system might not be restricted to their previously described antiviral and anti-retroelement activities. While it remains unclear whether the APOBEC3 proteins contribute to abundant genetic changes that accumulate in tumor cells, the recent findings suggest that at least some members of the family constitute a threat to cellular genomic integrity, and that their selection through evolution must have been governed by a cost-benefit balance.

Acknowledgments

We thank C. Lilley and B. Lamarche for helpful discussions. Work in the Weitzman lab was partially supported by NIH grants AI067952 and AI074967 (M.D.W.). This work was also funded by fellowships from the Instituto de Salud “Carlos III”/Consejo Superior de Investigaciones Científicas/Salk Institute and the Lynn Streim Postdoctoral Endowment Fellowship (I.N.), and the Natural Sciences and Engineering Research Council of Canada (S.L.).

References

  • Hamilton CE, Papavasiliou FN, Rosenberg BR. Diverse functions for DNA and RNA editing in the immune system. RNA Biol 2010; 7:220 - 228; PMID: 20220309; http://dx.doi.org/10.4161/rna.7.2.11344
  • Conticello SG, Langlois MA, Yang Z, Neuberger MS. DNA deamination in immunity: AID in the context of its APOBEC relatives. Adv Immunol 2007; 94:37 - 73; PMID: 17560271; http://dx.doi.org/10.1016/S0065-2776(06)94002-4
  • Di Noia JM, Neuberger MS. Molecular mechanisms of antibody somatic hypermutation. Annu Rev Biochem 2007; 76:1 - 22; PMID: 17328676; http://dx.doi.org/10.1146/annurev.biochem.76.061705.090740
  • Stavnezer J, Guikema JE, Schrader CE. Mechanism and regulation of class switch recombination. Annu Rev Immunol 2008; 26:261 - 292; PMID: 18370922; http://dx.doi.org/10.1146/annurev.immunol.26.021607.090248
  • Chiu YL, Greene WC. The APOBEC3 cytidine deaminases: an innate defensive network opposing exogenous retroviruses and endogenous retroelements. Annu Rev Immunol 2008; 26:317 - 353; PMID: 18304004; http://dx.doi.org/10.1146/annurev.immunol.26.021607.090350
  • Harris RS, Liddament MT. Retroviral restriction by APOBEC proteins. Nat Rev Immunol 2004; 4:868 - 877; PMID: 15516966; http://dx.doi.org/10.1038/nri1489
  • Holmes RK, Malim MH, Bishop KN. APOBEC-mediated viral restriction: not simply editing?. Trends Biochem Sci 2007; 32:118 - 128; PMID: 17303427; http://dx.doi.org/10.1016/j.tibs.2007.01.004
  • Harris RS, Bishop KN, Sheehy AM, Craig HM, Petersen-Mahrt SK, Watt IN, et al. DNA deamination mediates innate immunity to retroviral infection. Cell 2003; 113:803 - 809; PMID: 12809610; http://dx.doi.org/10.1016/S0092-8674(03)00423-9
  • Mangeat B, Turelli P, Caron G, Friedli M, Perrin L, Trono D. Broad antiretroviral defence by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature 2003; 424:99 - 103; PMID: 12808466; http://dx.doi.org/10.1038/nature01709
  • Zhang H, Yang B, Pomerantz RJ, Zhang C, Arunachalam SC, Gao L. The cytidine deaminase CEM15 induces hypermutation in newly synthesized HIV-1 DNA. Nature 2003; 424:94 - 98; PMID: 12808465; http://dx.doi.org/10.1038/nature01707
  • Lecossier D, Bouchonnet F, Clavel F, Hance AJ. Hypermutation of HIV-1 DNA in the absence of the Vif protein. Science 2003; 300:1112; PMID: 12750511; http://dx.doi.org/10.1126/science.1083338
  • Suspène R, Guétard D, Henry M, Sommer P, Wain-Hobson S, Vartanian JP. Extensive editing of both hepatitis B virus DNA strands by APOBEC3 cytidine deaminases in vitro and in vivo. Proc Natl Acad Sci USA 2005; 102:8321 - 8326; PMID: 15919829; http://dx.doi.org/10.1073/pnas.0408223102
  • Vartanian JP, Guetard D, Henry M, Wain-Hobson S. Evidence for editing of human papillomavirus DNA by APOBEC3 in benign and precancerous lesions. Science 2008; 320:230 - 233; PMID: 18403710; http://dx.doi.org/10.1126/science.1153201
  • Yang B, Chen K, Zhang C, Huang S, Zhang H. Virion-associated uracil DNA glycosylase-2 and apurinic/apyrimidinic endonuclease are involved in the degradation of APOBEC3G-edited nascent HIV-1 DNA. J Biol Chem 2007; 282:11667 - 11675; PMID: 17272283; http://dx.doi.org/10.1074/jbc.M606864200
  • Kaiser SM, Emerman M. Uracil DNA glycosylase is dispensable for human immunodeficiency virus type 1 replication and does not contribute to the antiviral effects of the cytidine deaminase Apobec3G. J Virol 2006; 80:875 - 882; PMID: 16378989; http://dx.doi.org/10.1128/JVI.80.2.875-82.2006
  • Mbisa JL, Barr R, Thomas JA, Vandegraaff N, Dorweiler IJ, Svarovskaia ES, et al. Human immunodeficiency virus type 1 cDNAs produced in the presence of APOBEC3G exhibit defects in plus-strand DNA transfer and integration. J Virol 2007; 81:7099 - 7110; PMID: 17428871; http://dx.doi.org/10.1128/JVI.00272-07
  • Schumacher AJ, Haché G, Macduff DA, Brown WL, Harris RS. The DNA deaminase activity of human APOBEC3G is required for Ty1, MusD and human immunodeficiency virus type 1 restriction. J Virol 2008; 82:2652 - 2660; PMID: 18184715; http://dx.doi.org/10.1128/JVI.02391-07
  • Stenglein MD, Burns MB, Li M, Lengyel J, Harris RS. APOBEC3 proteins mediate the clearance of foreign DNA from human cells. Nat Struct Mol Biol 2010; 17:222 - 229; PMID: 20062055; http://dx.doi.org/10.1038/nsmb.1744
  • Chen H, Lilley CE, Yu Q, Lee DV, Chou J, Narvaiza I, et al. APOBEC3A is a potent inhibitor of adeno-associated virus and retrotransposons. Curr Biol 2006; 16:480 - 485; PMID: 16527742; http://dx.doi.org/10.1016/j.cub.2006.01.031
  • Suspène R, Henry M, Guillot S, Wain-Hobson S, Vartanian JP. Recovery of APOBEC3-edited human immunodeficiency virus G→A hypermutants by differential DNA denaturation PCR. J Gen Virol 2005; 86:125 - 129; PMID: 15604439; http://dx.doi.org/10.1099/vir.0.80426-0
  • Rosenberg BR, Papavasiliou FN. Beyond SHM and CSR: AID and related cytidine deaminases in the host response to viral infection. Adv Immunol 2007; 94:215 - 244; PMID: 17560276; http://dx.doi.org/10.1016/S0065-2776(06)94007-3
  • Petit V, Vartanian JP, Wain-Hobson S. Powerful mutators lurking in the genome. Philos Trans Soc Lond B Biol Sci 2009; 364:705 - 715; PMID: 19042181; http://dx.doi.org/10.1098/rstb.2008.0272
  • Liu M, Schatz DG. Balancing AID and DNA repair during somatic hypermutation. Trends Immunol 2009; 30:173 - 181; PMID: 19303358; http://dx.doi.org/10.1016/j.it.2009.01.007
  • Nagaoka H, Tran TH, Kobayashi M, Aida M, Honjo T. Preventing AID, a physiological mutator, from deleterious activation: regulation of the genomic instability that is associated with antibody diversity. Int Immunol 2010; 22:227 - 235; PMID: 20207715; http://dx.doi.org/10.1093/intimm/dxq023
  • Liu M, Duke JL, Richter DJ, Vinuesa CG, Goodnow CC, Kleinstein SH, et al. Two levels of protection for the B cell genome during somatic hypermutation. Nature 2008; 451:841 - 845; PMID: 18273020; http://dx.doi.org/10.1038/nature06547
  • Staszewski O, Baker RE, Ucher AJ, Martier R, Stavnezer J, Guikema JE. Activation-induced cytidine deaminase induces reproducible DNA breaks at many non-Ig Loci in activated B cells. Mol Cell 2011; 41:232 - 242; PMID: 21255732; http://dx.doi.org/10.1016/j.molcel.2011.01.007
  • Tsai AG, Lu H, Raghavan SC, Muschen M, Hsieh CL, Lieber MR. Human chromosomal translocations at CpG sites and a theoretical basis for their lineage and stage specificity. Cell 2008; 135:1130 - 1142; PMID: 19070581; http://dx.doi.org/10.1016/j.cell.2008.10.035
  • Robbiani DF, Bunting S, Feldhahn N, Bothmer A, Camps J, Deroubaix S, et al. AID produces DNA double-strand breaks in non-Ig genes and mature B cell lymphomas with reciprocal chromosome translocations. Mol Cell 2009; 36:631 - 641; PMID: 19941823; http://dx.doi.org/10.1016/j.molcel.2009.11.007
  • Hasham MG, Donghia NM, Coffey E, Maynard J, Snow KJ, Ames J, et al. Widespread genomic breaks generated by activation-induced cytidine deaminase are prevented by homologous recombination. Nat Immunol 2010; 11:820 - 826; PMID: 20657597; http://dx.doi.org/10.1038/ni.1909
  • Wang M, Yang Z, Rada C, Neuberger MS. AID upmutants isolated using a high-throughput screen highlight the immunity/cancer balance limiting DNA deaminase activity. Nat Struct Mol Biol 2009; 16:769 - 776; PMID: 19543289; http://dx.doi.org/10.1038/nsmb.1623
  • Landry S, Narvaiza I, Linfesty DC, Weitzman MD. APOBEC3A can activate the DNA damage response and cause cell cycle arrest. EMBO Rep 2011; 12:444 - 450; PMID: 21460793; http://dx.doi.org/10.1038/embor.2011.46
  • Fernandez-Capetillo O, Celeste A, Nussenzweig A. Focusing on foci: H2AX and the recruitment of DNA-damage response factors. Cell Cycle 2003; 2:426 - 427; PMID: 12963833; http://dx.doi.org/10.4161/cc.2.5.509
  • Bransteitter R, Pham P, Scharff MD, Goodman MF. Activation-induced cytidine deaminase deaminates deoxycytidine on single-stranded DNA but requires the action of RNase. Proc Natl Acad Sci USA 2003; 100:4102 - 4107; PMID: 12651944; http://dx.doi.org/10.1073/pnas.0730835100
  • Suspène R, Sommer P, Henry M, Ferris S, Guetard D, Pochet S, et al. APOBEC3G is a single-stranded DNA cytidine deaminase and functions independently of HIV reverse transcriptase. Nucleic Acids Res 2004; 32:2421 - 2429; PMID: 15121899; http://dx.doi.org/10.1093/nar/gkh554
  • Yu Q, König R, Pillai S, Chiles K, Kearney M, Palmer S, et al. Single-strand specificity of APOBEC3G accounts for minus-strand deamination of the HIV genome. Nat Struct Mol Biol 2004; 11:435 - 442
  • Chaudhuri J, Tian M, Khuong C, Chua K, Pinaud E, Alt FW. Transcription-targeted DNA deamination by the AID antibody diversification enzyme. Nature 2003; 422:726 - 730; PMID: 12692563; http://dx.doi.org/10.1038/nature01574
  • Ramiro AR, Stavropoulos P, Jankovic M, Nussenzweig MC. Transcription enhances AID-mediated cytidine deamination by exposing single-stranded DNA on the nontemplate strand. Nat Immunol 2003; 4:452 - 456; PMID: 12692548; http://dx.doi.org/10.1038/ni920
  • Pham P, Bransteitter R, Petruska J, Goodman MF. Processive AID-catalysed cytosine deamination on single-stranded DNA simulates somatic hypermutation. Nature 2003; 424:103 - 107; PMID: 12819663; http://dx.doi.org/10.1038/nature01760
  • Shen HM, Poirier MG, Allen MJ, North J, Lal R, Widom J, et al. The activation-induced cytidine deaminase (AID) efficiently targets DNA in nucleosomes but only during transcription. J Exp Med 2009; 206:1057 - 1071; PMID: 19380635; http://dx.doi.org/10.1084/jem.20082678
  • Khobta A, Epe B. Interactions between DNA damage, repair and transcription. Mutat Res 2011; In press
  • Suspène R, Aynaud MM, Guetard D, Henry M, Eckhoff G, Marchio A, et al. Somatic hypermutation of human mitochondrial and nuclear DNA by APOBEC3 cytidine deaminases, a pathway for DNA catabolism. Proc Natl Acad Sci USA 2011; 108:4858 - 4863; PMID: 21368204; http://dx.doi.org/10.1073/pnas.1009687108
  • Imai K, Catalan N, Plebani A, Marodi L, Sanal O, Kumaki S, et al. Hyper-IgM syndrome type 4 with a B lymphocyte-intrinsic selective deficiency in Ig class-switch recombination. J Clin Invest 2003; 112:136 - 142; PMID: 12840068
  • Muramatsu M, Sankaranand VS, Anant S, Sugai M, Kinoshita K, Davidson NO, et al. Specific expression of activation-induced cytidine deaminase (AID), a novel member of the RNA-editing deaminase family in germinal center B cells. J Biol Chem 1999; 274:18470 - 18476; PMID: 10373455; http://dx.doi.org/10.1074/jbc.274.26.18470
  • Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, Shinkai Y, Honjo T. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell 2000; 102:553 - 563; PMID: 11007474; http://dx.doi.org/10.1016/S0092-8674(00)00078-7
  • Revy P, Muto T, Levy Y, Geissmann F, Plebani A, Sanal O, et al. Activation-induced cytidine deaminase (AID) deficiency causes the autosomal recessive form of the Hyper-IgM syndrome (HIGM2). Cell 2000; 102:565 - 575; PMID: 11007475; http://dx.doi.org/10.1016/S0092-8674(00)00079-9
  • Delker RK, Fugmann SD, Papavasiliou FN. A coming-of-age story: activation-induced cytidine deaminase turns 10. Nat Immunol 2009; 10:1147 - 1153; PMID: 19841648; http://dx.doi.org/10.1038/ni.1799
  • Crouch EE, Li Z, Takizawa M, Fichtner-Feigl S, Gourzi P, Montano C, et al. Regulation of AID expression in the immune response. J Exp Med 2007; 204:1145 - 1156; PMID: 17452520; http://dx.doi.org/10.1084/jem.20061952
  • Dorsett Y, McBride KM, Jankovic M, Gazumyan A, Thai TH, Robbiani DF, et al. MicroRNA-155 suppresses activation-induced cytidine deaminase-mediated Myc-Igh translocation. Immunity 2008; 28:630 - 638; PMID: 18455451; http://dx.doi.org/10.1016/j.immuni.2008.04.002
  • Patenaude AM, Orthwein A, Hu Y, Campo VA, Kavli B, Buschiazzo A, et al. Active nuclear import and cytoplasmic retention of activation-induced deaminase. Nat Struct Mol Biol 2009; 16:517 - 527; PMID: 19412186; http://dx.doi.org/10.1038/nsmb.1598
  • Pasqualucci L, Kitaura Y, Gu H, Dalla-Favera R. PKA-mediated phosphorylation regulates the function of activation-induced deaminase (AID) in B cells. Proc Natl Acad Sci USA 2006; 103:395 - 400; PMID: 16387847; http://dx.doi.org/10.1073/pnas.0509969103
  • Basu U, Franklin A, Alt FW. Post-translational regulation of activation-induced cytidine deaminase. Philos Trans R Soc Lond B Biol Sci 2009; 364:667 - 673; PMID: 19010772; http://dx.doi.org/10.1098/rstb.2008.0194
  • Aoufouchi S, Faili A, Zober C, D'Orlando O, Weller S, Weill JC, et al. Proteasomal degradation restricts the nuclear lifespan of AID. J Exp Med 2008; 205:1357 - 1368; PMID: 18474627; http://dx.doi.org/10.1084/jem.20070950
  • Xu Z, Fulop Z, Wu G, Pone EJ, Zhang J, Mai T, et al. 14-3-3 adaptor proteins recruit AID to 5′-AGCT-3′-rich switch regions for class switch recombination. Nat Struct Mol Biol 2010; 17:1124 - 1135; PMID: 20729863; http://dx.doi.org/10.1038/nsmb.1884
  • Orthwein A, Patenaude AM, Affar el B, Lamarre A, Young JC, Di Noia JM. Regulation of activation-induced deaminase stability and antibody gene diversification by Hsp90. J Exp Med 2010; 207:2751 - 2765; PMID: 21041454; http://dx.doi.org/10.1084/jem.20101321
  • Storck S, Aoufouchi S, Weill JC, Reynaud CA. AID and partners: for better and (not) for worse. Curr Opin Immunol 2011; 23:337 - 344; PMID: 21439803; http://dx.doi.org/10.1016/j.coi.2011.02.002
  • Okazaki IM, Kotani A, Honjo T. Role of AID in tumorigenesis. Adv Immunol 2007; 94:245 - 273; PMID: 17560277; http://dx.doi.org/10.1016/S0065-2776(06)94008-5
  • Marusawa H, Takai A, Chiba T. Role of activation-induced cytidine deaminase in inflammation-associated cancer development. Adv Immunol 2011; 111:109 - 141; PMID: 21970953; http://dx.doi.org/10.1016/B978-0-12-385991-4.00003-9
  • Peng G, Lei KJ, Jin W, Greenwell-Wild T, Wahl SM. Induction of APOBEC3 family proteins, a defensive maneuver underlying interferon-induced anti-HIV-1 activity. J Exp Med 2006; 203:41 - 46; PMID: 16418394; http://dx.doi.org/10.1084/jem.20051512
  • Bonvin M, Achermann F, Greeve I, Stroka D, Keogh A, Inderbitzin D, et al. Interferon-inducible expression of APOBEC3 editing enzymes in human hepatocytes and inhibition of hepatitis B virus replication. Hepatology 2006; 43:1364 - 1374; PMID: 16729314; http://dx.doi.org/10.1002/hep.21187
  • Stopak KS, Chiu YL, Kropp J, Grant RM, Greene WC. Distinct patterns of cytokine regulation of APOBEC3G expression and activity in primary lymphocytes, macrophages and dendritic cells. J Biol Chem 2007; 282:3539 - 3546; PMID: 17110377; http://dx.doi.org/10.1074/jbc.M610138200
  • Koning FA, Newman EN, Kim EY, Kunstman KJ, Wolinsky SM, Malim MH. Defining APOBEC3 expression patterns in human tissues and hematopoietic cell subsets. J Virol 2009; 83:9474 - 9485; PMID: 19587057; http://dx.doi.org/10.1128/JVI.01089-09
  • Pauklin S, Sernandez IV, Bachmann G, Ramiro AR, Petersen-Mahrt SK. Estrogen directly activates AID transcription and function. J Exp Med 2009; 206:99 - 111; PMID: 19139166; http://dx.doi.org/10.1084/jem.20080521
  • Refsland EW, Stenglein MD, Shindo K, Albin JS, Brown WL, Harris RS. Quantitative profiling of the full APOBEC3 mRNA repertoire in lymphocytes and tissues: implications for HIV-1 restriction. Nucleic Acids Res 2010; 38:4274 - 4284; PMID: 20308164; http://dx.doi.org/10.1093/nar/gkq174
  • Rose KM, Marin M, Kozak SL, Kabat D. Transcriptional regulation of APOBEC3G, a cytidine deaminase that hypermutates human immunodeficiency virus. J Biol Chem 2004; 279:41744 - 41749; PMID: 15297452; http://dx.doi.org/10.1074/jbc.M406760200
  • Sarkis PT, Ying S, Xu R, Yu XF. STAT1-independent cell type-specific regulation of antiviral APOBEC3G by IFN-alpha. J Immunol 2006; 177:4530 - 4540; PMID: 16982890
  • Muckenfuss H, Kaiser JK, Krebil E, Battenberg M, Schwer C, Cichutek K, et al. Sp1 and Sp3 regulate basal transcription of the human APOBEC3G gene. Nucleic Acids Res 2007; 35:3784 - 3796; PMID: 17517765; http://dx.doi.org/10.1093/nar/gkm340
  • Farrow MA, Kim EY, Wolinsky SM, Sheehy AM. NFAT and IRF proteins regulate transcription of the anti-HIV gene, APOBEC3G. J Biol Chem 2011; 286:2567 - 2577; PMID: 21078663; http://dx.doi.org/10.1074/jbc.M110.154377
  • Gallois-Montbrun S, Kramer B, Swanson CM, Byers H, Lynham S, Ward M, et al. Antiviral protein APOBEC3G localizes to ribonucleoprotein complexes found in P bodies and stress granules. J Virol 2007; 81:2165 - 2178; PMID: 17166910; http://dx.doi.org/10.1128/JVI.02287-06
  • Huang J, Liang Z, Yang B, Tian H, Ma J, Zhang H. Derepression of MicroRNA-mediated Protein Translation Inhibition by Apolipoprotein B mRNA-editing Enzyme Catalytic Polypeptide-like 3G (APOBEC3G) and Its Family Members. J Biol Chem 2007; 282:33632 - 33640; PMID: 17848567; http://dx.doi.org/10.1074/jbc.M705116200
  • Huthoff H, Autore F, Gallois-Montbrun S, Fraternali F, Malim MH. RNA-dependent oligomerization of APOBEC3G is required for restriction of HIV-1. PLoS Pathog 2009; 5:1000330; PMID: 19266078; http://dx.doi.org/10.1371/journal.ppat.1000330
  • Iwatani Y, Takeuchi H, Strebel K, Levin JG. Biochemical activities of highly purified, catalytically active human APOBEC3G: correlation with antiviral effect. J Virol 2006; 80:5992 - 6002; PMID: 16731938; http://dx.doi.org/10.1128/JVI.02680-05
  • Kozak SL, Marin M, Rose KM, Bystrom C, Kabat D. The anti-HIV-1 editing enzyme APOBEC3G binds HIV-1 RNA and messenger RNAs that shuttle between polysomes and stress granules. J Biol Chem 2006; 281:29105 - 29119; PMID: 16887808; http://dx.doi.org/10.1074/jbc.M601901200
  • Kreisberg JF, Yonemoto W, Greene WC. Endogenous factors enhance HIV infection of tissue naive CD4 T cells by stimulating high molecular mass APOBEC3G complex formation. J Exp Med 2006; 203:865 - 870; PMID: 16606671; http://dx.doi.org/10.1084/jem.20051856
  • Shirakawa K, Takaori-Kondo A, Yokoyama M, Izumi T, Matsui M, Io K, et al. Phosphorylation of APOBEC3G by protein kinase A regulates its interaction with HIV-1 Vif. Nat Struct Mol Biol 2008; 15:1184 - 1191
  • Stenglein MD, Matsuo H, Harris RS. Two regions within the amino-terminal half of APOBEC3G cooperate to determine cytoplasmic localization. J Virol 2008; 82:9591 - 9599; PMID: 18667511; http://dx.doi.org/10.1128/JVI.02471-07
  • Chiu YL, Witkowska HE, Hall SC, Santiago M, Soros VB, Esnault C, et al. High-molecular-mass APOBEC3G complexes restrict Alu retrotransposition. Proc Natl Acad Sci USA 2006; 103:15588 - 15593; PMID: 17030807; http://dx.doi.org/10.1073/pnas.0604524103
  • Bennett RP, Presnyak V, Wedekind JE, Smith HC. Nuclear Exclusion of the HIV-1 host defense factor APOBEC3G requires a novel cytoplasmic retention signal and is not dependent on RNA binding. J Biol Chem 2008; 283:7320 - 7327; PMID: 18165230; http://dx.doi.org/10.1074/jbc.M708567200
  • McDougall WM, Smith HC. Direct evidence that RNA inhibits APOBEC3G ssDNA cytidine deaminase activity. Biochem Biophys Res Commun 2011; 412:612 - 617; PMID: 21856286; http://dx.doi.org/10.1016/j.bbrc.2011.08.009
  • Thielen BK, Klein KC, Walker LW, Rieck M, Buckner JH, Tomblingson GW, et al. T cells contain an RNase-insensitive inhibitor of APOBEC3G deaminase activity. PLoS Pathog 2007; 3:1320 - 1334; PMID: 17892323; http://dx.doi.org/10.1371/journal.ppat.0030135
  • Wichroski MJ, Robb GB, Rana TM. Human retroviral host restriction factors APOBEC3G and APOBEC3F localize to mRNA processing bodies. PLoS Pathog 2006; 2:41; PMID: 16699599; http://dx.doi.org/10.1371/journal.ppat.0020041
  • Bogerd HP, Wiegand HL, Hulme AE, Garcia-Perez JL, O'Shea KS, Moran JV, et al. Cellular inhibitors of long interspersed element 1 and Alu retrotransposition. Proc Natl Acad Sci USA 2006; 103:8780 - 8785; PMID: 16728505; http://dx.doi.org/10.1073/pnas.0603313103
  • Niewiadomska AM, Tian C, Tan L, Wang T, Sarkis PTN, Yu XF. Differential inhibition of long interspersed element 1 by APOBEC3 does not correlate with high-molecular-mass-complex formation or P-body association. J Virol 2007; 81:9577 - 9583; PMID: 17582006; http://dx.doi.org/10.1128/JVI.02800-06
  • Mehta A, Kinter MT, Sherman NE, Driscoll DM. Molecular cloning of apobec-1 complementation factor, a novel RNA-binding protein involved in the editing of apolipoprotein B mRNA. Mol Cell Biol 2000; 20:1846 - 1854; PMID: 10669759; http://dx.doi.org/10.1128/MCB.20.5.1846-54.2000
  • Lellek H, Kirsten R, Diehl I, Apostel F, Buck F, Greeve J. Purification and molecular cloning of a novel essential component of the apolipoprotein B mRNA editing enzyme-complex. J Biol Chem 2000; 275:19848 - 19856; PMID: 10781591; http://dx.doi.org/10.1074/jbc.M001786200
  • Zhang W, Zhang X, Tian C, Wang T, Sarkis PTN, Fang Y, et al. Cytidine deaminase APOBEC3B interacts with heterogeneous nuclear ribonucleoprotein K and suppresses hepatitis B virus expression. Cell Microbiol 2008; 10:112 - 121; PMID: 17672864
  • Smith HC, Bennett RP, Kizilyer A, McDougall WM, Prohaska KM. Functions and regulation of the APOBEC family of proteins. Semin Cell Dev Biol 2011; In press
  • Narvaiza I, Linfesty DC, Greener BN, Hakata Y, Pintel DJ, Logue E, et al. Deaminase-independent inhibition of parvoviruses by the APOBEC3A cytidine deaminase. PLoS Pathog 2009; 5:1000439; PMID: 19461882; http://dx.doi.org/10.1371/journal.ppat.1000439
  • Thielen BK, McNevin JP, McElrath MJ, Hunt BVS, Klein KC, Lingappa JR. Innate immune signaling induces high levels of TC-specific deaminase activity in primary monocyte-derived cells through expression of APOBEC3A isoforms. J Biol Chem 2010; 285:27753 - 27766; PMID: 20615867; http://dx.doi.org/10.1074/jbc.M110.102822
  • Bulliard Y, Narvaiza I, Bertero A, Peddi S, Rohrig UF, Ortiz M, et al. Structure-function analyses point to a polynucleotide-accommodating groove essential for APOBEC3A restriction activities. J Virol 2011; 85:1765 - 1776; PMID: 21123384; http://dx.doi.org/10.1128/JVI.01651-10
  • Sweasy JB, Lang T, DiMaio D. Is base excision repair a tumor suppressor mechanism?. Cell Cycle 2006; 5:250 - 259; PMID: 16418580; http://dx.doi.org/10.4161/cc.5.3.2414
  • Berger G, Durand S, Fargier G, Nguyen XN, Cordeil S, Bouaziz S, et al. APOBEC3A Is a Specific Inhibitor of the Early Phases of HIV-1 Infection in Myeloid Cells. PLoS Pathog 2011; 7:1002221; PMID: 21966267; http://dx.doi.org/10.1371/journal.ppat.1002221
  • Sawyer SL, Emerman M, Malik HS. Ancient adaptive evolution of the primate antiviral DNA-editing enzyme APOBEC3G. PLoS Biol 2004; 2:275; PMID: 15269786; http://dx.doi.org/10.1371/journal.pbio.0020275
  • Kidd JM, Newman TL, Tuzun E, Kaul R, Eichler EE. Population stratification of a common APOBEC gene deletion polymorphism. PLoS Genet 2007; 3:63; PMID: 17447845; http://dx.doi.org/10.1371/journal.pgen.0030063
  • OhAinle M, Kerns JA, Li MM, Malik HS, Emerman M. Antiretroelement activity of APOBEC3H was lost twice in recent human evolution. Cell Host Microbe 2008; 4:249 - 259; PMID: 18779051; http://dx.doi.org/10.1016/j.chom.2008.07.005
  • Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 2005; 436:1186 - 1190; PMID: 15995699; http://dx.doi.org/10.1038/nature03884
  • Gasser S, Raulet DH. The DNA damage response arouses the immune system. Cancer Res 2006; 66:3959 - 3962; PMID: 16618710; http://dx.doi.org/10.1158/0008-5472.CAN-05-4603
  • Gourzi P, Leonova T, Papavasiliou FN. A role for activation-induced cytidine deaminase in the host response against a transforming retrovirus. Immunity 2006; 24:779 - 786; PMID: 16782033; http://dx.doi.org/10.1016/j.immuni.2006.03.021
  • Norman JM, Mashiba M, McNamara LA, Onafuwa-Nuga A, Chiari-Fort E, Shen W, et al. The antiviral factor APOBEC3G enhances the recognition of HIV-infected primary T cells by natural killer cells. Nat Immunol 2011; 12:975 - 983; PMID: 21874023; http://dx.doi.org/10.1038/ni.2087
  • Zaheen A, Boulianne B, Parsa JY, Ramachandran S, Gommerman JL, Martin A. AID constrains germinal center size by rendering B cells susceptible to apoptosis. Blood 2009; 114:547 - 554; PMID: 19478044; http://dx.doi.org/10.1182/blood-2009-03-211763
  • Barré B, Coqueret O, Perkins ND. Regulation of activity and function of the p52 NFkappaB subunit following DNA damage. Cell Cycle 2010; 9:4795 - 4804; PMID: 21131783; http://dx.doi.org/10.4161/cc.9.24.14245
  • Xu R, Zhang X, Zhang W, Fang Y, Zheng S, Yu XF. Association of human APOBEC3 cytidine deaminases with the generation of hepatitis virus B x antigen mutants and hepatocellular carcinoma. Hepatology 2007; 46:1810 - 1820; PMID: 17847074; http://dx.doi.org/10.1002/hep.21893
  • Komatsu A, Nagasaki K, Fujimori M, Amano J, Miki Y. Identification of novel deletion polymorphisms in breast cancer. Int J Oncol 2008; 33:261 - 270
  • Ding Q, Chang CJ, Xie X, Xia W, Yang JY, Wang SC, et al. APOBEC3G promotes liver metastasis in an orthotopic mouse model of colorectal cancer and predicts human hepatic metastasis. J Clin Invest 2011; 121:4526 - 4536
  • Fritz EL, Papavasiliou FN. Cytidine deaminases: AIDing DNA demethylation?. Genes Dev 2010; 24:2107 - 2114
  • Guo JU, Su Y, Zhong C, Ming GL, Song H. Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain. Cell 2011; 145:423 - 434; PMID: 21496894; http://dx.doi.org/10.1016/j.cell.2011.03.022

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.