Zebrafish as a Model System to Study DNA Damage and Repair

download Zebrafish as a Model System to Study DNA Damage and Repair

of 9

Transcript of Zebrafish as a Model System to Study DNA Damage and Repair

  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    1/9

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003mailto:[email protected]:[email protected]://www.elsevier.com/locate/mutreshttp://www.elsevier.com/locate/molmuthttp://www.sciencedirect.com/science/journal/00275107http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    2/9

    Please cite this article in pressas: D.-S. Pei, P.R. Strauss, Zebrafish as a model system to study DNA damage and repair, Mutat. Res.: Fundam. Mol.

    Mech. Mutagen. (2012), http://dx.doi.org/10.1016/j.mrfmmm.2012.10.003

    ARTICLE IN PRESSGModel

    MUT-11229; No.of Pages9

    2 D.-S.Pei, P.R. Strauss / Mutation Researchxxx (2012) xxxxxx

    and repair pathways. First, zebrafish genomic DNA contains ortho-

    logues of genes involved in all the DNA repair pathways in higher

    eukaryotes (Table 1). Second, it is easy to perform morpholino-

    based [6,7] or shRNA knockdown [8,9] studies to directly address

    the role of specific DNA damage response genes in each repair

    pathway. Third, a new mutagenesis technology involving locus-

    specific zinc-finger nucleases (ZFNs) and transcription activator

    like effector (TALE) proteins uses the NHEJ pathway to manipulate

    the zebrafish genome genetically [1015]. ZFNs and TALEs are a

    fusion between the cleavage domain of theFokIrestriction enzyme

    and zinc-finger motifs or transcriptionalactivators designed to rec-

    ognize a specific DNAtarget sequence. They can introduce genomic

    lesions at a specific site to stimulate NHEJ-mediated repair of tar-

    geted double strand breaks (DSBs) and thus generate mutations in

    selected DNArepair genes.Therefore,the role of specific DNArepair

    genes can be addressed directly. Moreover, casper transparent

    zebrafish mutants are an asset to both toxicology and carcinogene-

    sis studies for monitoring damage-induced phenotypes from later

    development to adult stages [16]. These strategies are helpful to

    explore whether selected gene products in different DNA repair

    pathways may playadditional rolesin embryological development.

    This review focuses on recent work on DNA damage and repair and

    thesubsequent DNAdamage response (DDR) in zebrafish with spe-

    cial emphasis on base excision repair (BER) in early embryological

    development.

    2. DNA-repair pathways studied in zebrafish

    Although zebrafish have proved usefulin toxicologyand embry-

    onic development studies for some time, they have not been used

    to their fullest potential in studies of DNA damage and repair.

    While many zebrafish studies focus solely on cytotoxicity or p53-

    mediated response to DNA damage, the outcomes are ultimately

    the results of specific DNA damage and the related repair path-

    ways.These pathwaysinclude direct reversal (DR),mismatchrepair

    (MMR), nucleotide excision repair (NER), BER, non-homologous

    end joining (NHEJ), and homologous recombination (HR) pathway

    (Tables1and2). Note that DSB, where both strands of theDNA helix

    are disrupted, are repaired by NHEJ or HR, depending on the nature

    of the break and the stage of the cell cycle. Each pathway involves

    recognition of the specific damage, damage removal, which may

    involve cleavage of one or both strands, resynthesis and ligation of

    the repaired strand(s). While the mechanisms in each pathway are

    distinct, there is some overlap among pathways.

    2.1. Direct reversal

    The DR pathway does not involve breakage of the phosphodi-

    ester backbone but instead reverses the damage specifically and

    directly [17]. The prototype enzyme for DR is methyl guanine

    methyl transferase (Mgmt), which reverses alkylation of gua-nine. Although the zebrafish genome contains mgmt, no studies

    in zebrafish have been published at this time. However, a second

    system of particular physiological significance for fish is pho-

    toenzymatic repair (PER), which reverses the formation of UV-B

    (315280nm) or UV-C (280100nm)-induced adjacent cyclopy-

    rimidine dimers ( CPD). PER is carried out by a single enzyme,

    deoxyribodipyrimidine photolyase, encoded byphr. Notably, PERis

    not found in placental mammals, which rely solely on the less effi-

    cientNER[18]. PERis activatedby irradiation with long wavelength

    (300500nm) UVA/visible light [1921]. Fidelity of photo repair

    has been evaluated in zebrafish embryos exposed to UVB followed

    byUVA exposure to repairCPD [19,2224]. Notethat oxidativedam-

    age resulting from exposure to ultraviolet light is repaired by base

    excision repair (see below) [18,22,23,2528] .

    2.2. Nucleotide excision repair

    NER, found in all organisms including fish [29], excises and

    repairs pyrimidine dimers. There are two NER pathways distin-

    guished by whether the lesion is on the transcribed strand of an

    active gene (TCR) or the rest of the genome (GGR) [30]. There are

    no studies at this time that distinguish TCR from GGR in zebrafish,

    although the presence of the PER system could conceivably alter

    thebalance between TCRand GGR. Other studies relate primarily to

    dose dependence of exposure of whole zebrafish or zebrafish liver

    cells [31,32]toUV andthe responseto UV exposureof various genes

    involved in NER such as cdkI (cyclin-dependent kinase inhibitor),

    xpc, ddb2, p53, gadd45a and cyclinG1 [25,29,3336]. Interestingly,

    after exposure to wastewater effluent, adult male zebrafish and

    zebrafish liver cells demonstrated altered xpcandxpa expressions

    [37]. Alterations depended on the source ofwaste water and the

    time of year.xpd from zebrafish wasclonedand theexpression pat-

    tern in different tissues and different stages was investigated [38].

    Whilexpd/ercc2 was expressed in all developmental stages includ-

    ing unfertilized eggs, [38], expression ofxpa could not be detected

    by western blot analysis in early stage embryos prior t o hatching,

    even after UV irradiation [24,39,40].

    Exposure to the potent synthetic estrogen, 17alpha-

    ethinylestradiol (EE2) caused significant decreases in several

    hepatic NER genes, indicating potential decreased NER capacity

    and presumably increased cancer risk. In fact, exposure of cultured

    zebrafish liver cells to physiologically relevant concentrations

    of EE2 reduced the ability to repair a damaged reporter plasmid

    [35].

    2.3. Base excision repair

    BER recognizes non-bulky lesions such as 8-oxoguanine (8-

    oxoG) and the presence of uracil or abasic (AP sites) (Fig. 1). The

    lesion is recognized by a glycosylase that removes the inappropri-

    ate base without cleaving the backbone and generates an AP site

    (Fig. 1, Reaction 1). The AP site is then cleaved 5 to the deoxyri-

    bosephosphate by AP endonuclease 1 (Apex1), generating the free3 hydroxyl group required for the repair polymerase (Fig. 1, Reac-

    tion 2). The dangling dRP is removed by DNA polymerase (PolB),

    which fills the gap (Fig. 1, Reactions 3a and 4a). Some glycosylases

    not only remove the offending base but also nick the AP site on

    the 3 side, leaving a blocking lesion for PolB. In that case, Apex1 is

    required to trim the dRP, which results in a single nucleotide gap

    that is filled by PolB after which a ligase seals the nick (Fig. 1, Reac-

    tion 5a). On occasion, chemical modification prevents removal of

    the dRP. Under those conditions, PCNA and one of the replicative

    polymerases (or possibly PolB) displaces the downstream strand

    and inserts up to 5 nucleotides (Fig. 1, Reaction 3b). The displaced

    strand is cleaved by flap endonuclease1 (Fen1) (Fig. 1, Reaction 4b)

    and a ligase seals the nick (Fig. 1, Reaction 5b). Accessory proteins

    include Xrcc1 and Parp. Biochemical studies of zebrafish enzymesinvolved in BER include PolB [41] and Apex1 [26]. Like Xpa, PolB

    is missing in early stage zebrafish embryos [28] so that before 13h

    post fertilization (hpf), BER in early stage embryos relies on one or

    more aphidicolin-sensitive DNApolymerase(s) [28]. Detailed stud-

    ies of pathway regulation as studied in zebrafish and in mice are

    presented in Section 3 below.

    Toxicity studies of agents that result in oxidative damage to

    DNA that are likely to be repaired by BER in zebrafish embryos

    or adults include those involving chronic oxidation from expo-

    sure to copper [33], and the pyrethroid insecticide cypermethrin

    [4244]. Chronic exposure to the former resulted in decreased

    apex1 expression compared to control, while exposure to cyper-

    methrin down-regulated ogg1 gene expression, induced oxidative

    stress and upregulated antioxidant proteins (Cu/Zn-dependent

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    3/9

    Please cite this article in pressas: D.-S. Pei, P.R. Strauss, Zebrafish as a model system to study DNA damage and repair, Mutat. Res.: Fundam. Mol.

    Mech. Mutagen. (2012), http://dx.doi.org/10.1016/j.mrfmmm.2012.10.003

    ARTICLE IN PRESSGModel

    MUT-11229; No.of Pages9

    D.-S. Pei,P.R. Strauss / Mutation Researchxxx (2012) xxxxxx 3

    Table 1

    The major genesof differentDNA repair pathways in zebrafish.

    Types of DNA repair Major genes Accession ID Protein size (aa)

    Direct reversal mgmt XP 684479.2 187

    phr NP 957358.1 516

    Base excision repair ung NP 957268.1 291

    ogg1 NP 001074145.1 268

    apex1 (S, L)a NP 998586.1 310

    parp1 (S) NP 001038407.1 1013

    polb (S) NP 001003879 337

    xrcc1 (S) NP 001003988.1 615

    lig3 (S) NP 001025345.1 752

    pol (L) NP 001034899.1 1105

    pol (L) XP 001920422.1 2288

    fen1 (L) NP 942115 330

    lig1(L) XP 685041.2 1058

    Mismatch repair msh2 NP 998689.1 936

    msh3 NP 001103184.1 1083

    msh6 NP 878280.1 1369

    mlh1 NP 956953.1 724

    pms2 XP 693648.3 851

    exo1 NP 998634.1 807

    pol NP 001034899.1 1105

    Nucleotide excision repair RNApolII (TCR)b XP 682682.1 1972

    csa/ercc8 (TCR) NP 001005984.1 400

    csb/ercc6 (TCR) XP 688972.2 1390rpa (TCR,GGR)c NP 956105.2 601

    xpa (TCR, GGR) NP 956765.1 549

    xpg(TCR, GGR) NP 001014337.1 249

    ercc1 (TCR,GGR) NP 001096608.1 342

    xpc NP 001038675.1 879

    xpe NP 956920.1 897

    hr23b NP 956858.1 382

    ddb2 NP 001076530 497

    Nonhomologous end joining ku70 NP 956198.1 409

    ku80 NP 001017360.1 727

    DNA-PK/prkdc XP 001919588.1 4119

    xrcc4 NP 957080.1 357

    lig4 NP 001096593.1 909

    artemis NP 001038566.1 639

    Homologous recombination atm XP 001334561.2 323

    rad51 AAH62849.1 338rad52 NP 001019622.1 409

    rad54 NP 957438.1 738

    pol NP 001034899.1 1105

    pol XP 001920422.1 2288

    lig1 XP 685041.2 1058

    NHEJ and HR rad50 XP 696859.3 1332

    mre11 NP 001001407.1 619

    nbn NP 001014819.1 818

    Translesion synthesis poleta NP 001035337.1 743

    poliota NP 001017834.1 710

    polkappa XP 691219.4 902

    pollambda NP 998408.1 566

    polmu NP 956542.1 507

    polnu NP 001093496.1 1146

    rev1 NP 001116772.1 1268

    polzeta XP 002665692.2 1615

    p53-Mediated Surveillance p53 NP 571402.1 373

    mdm2 NP 571439.2 475

    atm XP 002664603.2 2451

    atr XP 696163.4 2643

    p21 XP 003200962 170

    survivin (birc5a) NP 919378.1 142

    rad1 NP 957192.1 279

    rad9 XP 692358.2 402

    hus1 NP 001082965.1 284

    baxa NP 571637.1 192

    noxa (pmaip1) NP 001038939 45

    puma (bbc3) NP 001038937.1 181

    bcl2 NP 001025424.1 228

    bcl-xl NP 571882.1 238

    casp2 NP 001036160.1 435

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    4/9

    Please cite this article in pressas: D.-S. Pei, P.R. Strauss, Zebrafish as a model system to study DNA damage and repair, Mutat. Res.: Fundam. Mol.

    Mech. Mutagen. (2012), http://dx.doi.org/10.1016/j.mrfmmm.2012.10.003

    ARTICLE IN PRESSGModel

    MUT-11229; No.of Pages9

    4 D.-S.Pei, P.R. Strauss / Mutation Researchxxx (2012) xxxxxx

    Table 1 (Continued)

    Types of DNA repair Major genes Accession ID Protein size (aa)

    casp 3a NP 571952.1 282

    casp 6 NP 001018333.1 298

    casp 7 NP 001018443.1 316

    nur77 (nr4a1) NP 001002173.1 574

    p73 NP 899183.1 640

    a S short patch, L longpatch.b TCR transcription coupled repair.c GGR global genomic repair.

    superoxide dismutase, Mn-dependent superoxide dismutase, cata-

    lase, and glutathione peroxidase), resulting in apoptosis with

    caspase activation in zebrafish embryos.

    2.4. Mismatch repair

    Like BER, MMR repairs non-bulky lesions in DNA. However,

    the use of the word lesion is a misnomer, because the pathway

    is limited to recognition and repair of mispaired but undamaged

    bases. For that reason it plays a key role in maintaining genomic

    stability and suppressing homologous recombination. Defects in

    MMRare associated with predisposition to colon cancer including

    Hereditary Non-Polyposis Colorectal Cancer (HNPCC), resistance

    to certain chemotherapeutic agents, abnormalities in meiosis and

    male sterility. These outcomes have all been studied in zebrafish

    systems. Some of the zebrafish genes involved in MMR, notably

    the damage recognition proteins Msh6, Msh2, and Mlh1 [45,46],

    have been cloned and both temporal and spatial distribution

    have been followed [45,46]. However, zebrafish mlh3, replication

    Table 2

    Recent studies on DNA repair pathwaysin zebrafish.

    DNA repair pathway Topic Genes involved Reference

    DR UV-damaged-DNA binding activity in zebrafish [24]

    Photobiological effects of UV pho [22]

    UVA-induced photo recovery pho [23]

    BER Apex1 affects heart,brain and blood development apex1 [26]

    Copper exposure in zebrafish apex1 [33]

    BER in early zebrafish development apex1, polb [27]

    A novel regulatory circuit in BER pathway apex1, creb1, polb [28]

    Overexpression of Polb in E. coli polb [41]

    Cypermethrin exposure in zebrafish ogg1, p53 [44]

    NER 17-Ethinylestradiol affects NER genes expression xpc, xpd, xpa, xpf [34]UVC and oxidativedamage in cultured fish cells [25]

    17-Ethinylestradiol hinders NER in zebrafish liver cells xpc, xpa [35]p53-dependent NER pathway in zebrafish p53 [29]

    Time-course expression of DNA repair-related genes by UVB p53, xpc, ddb2, gadd45a, cyclinG1 [33]

    Wastewater treatment alters NER in zebrafish xpc,xpa [37]

    Cloning ofxpd in zebrafish xpd [38]

    1,10-phenanthroline (OP) stimulated UV damaged DNA binding proteins vitellogenin [39]

    UVR exposure in fish embryos [40]

    EE2 modulatesp53 but not NER pathways p53, p21, gadd45a [36]

    MMR MMR-dependent chromosomal instability from alkylation damage msh6 [53]

    Mlh1 deficiency for zebrafish male sterility mlh1 [48]

    Completion of meiosis lack ofmlh1 in zebrafish mlh1 [49]

    MMRdeficiency does not enhance ENU mutagenesis msh6 [51]

    Mutations in MMRdevelop tumors mlh1, mlh2, mlh6 [50]

    Cadmium down-regulates msh6 msh6 [68]

    Cloning ofmsh2 in zebrafish msh2 [46]

    Cloning ofmsh6 in zebrafish msh6 [45]

    Zebrafish: chiasmataand interference mlh1 [47]NHEJ SB transpositional recombination by NHEJ pathway dna-pk [69]

    Ionizing radiation-induced DNA damage. xrcc5/ku80 [54]

    Ku70 protects nervous system from radiation damage xrcc6/ku70 [55]

    The role of NHEJ in transgene concatemer formation [57]

    ZFN targeted disruption in zebrafish [11]

    ZFN minireview [10]

    ZFN targeted disruption in zebrafish [14]

    ZFNas gene therapy agents [70]

    NHEJ involved in LINE retrotransposition zfl2-2, ku70, art, ligIV [56]

    Rapidmutation of zebrafishgene by ZFNOPEN [13]

    Targeted mutagenesis using customized ZFN [12]

    Improved somatic mutagenesis in zebrafish using TALENs [15]

    HR Chi-stimulated HR and i ts application in zebrafish [71]

    HRand NHEJ in zebrafish [59], [58]

    Use of RecA fusion proteins for zebrafish genomic modifications reca [62]

    Rad52 and RPA mediated mutagenesis rad52, rpa [61]

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    5/9

    Please cite this article in pressas: D.-S. Pei, P.R. Strauss, Zebrafish as a model system to study DNA damage and repair, Mutat. Res.: Fundam. Mol.

    Mech. Mutagen. (2012), http://dx.doi.org/10.1016/j.mrfmmm.2012.10.003

    ARTICLE IN PRESSGModel

    MUT-11229; No.of Pages9

    D.-S. Pei,P.R. Strauss / Mutation Researchxxx (2012) xxxxxx 5

    Fig. 1. DNA base excision repairpathway (modified from Fortier et al. [23]). DNA base excision repairpathway. The step numberis indicated at the left in Arabic numerals.

    Steps 3a, 4a, and 5a occur in short patch (single-nucleotide) repair. Steps 3b, 4b, and 5b occur in long patch repair (insertion of two to six additional nucleotides). Step 3b

    uses the replicative DNA polymerase-, -, and -, in the presence of flap endonuclease 1 (FEN1) and PCNA (not shown).

    factor c (rfc) and pms2 remain to be explored. Expression ofmsh6

    relative to msh2 and the tissue distribution ofmsh2 varied with

    the stage of development [45] until the relative levels stabilizedat 120 hpf [46]. msh2 tended to be localized in the brain, eyes,

    telencephalon, and the fourth ventricle at 12 days post fertilization

    (dpf)-48 dpf embryos [46]. On theotherhand, Mlh1 foci detected by

    immunofluorescence tended to be in the distal regions of zebrafish

    synaptonemal complexes (SCs) [47].

    MMR is critical for sperm maturation. Male zebrafish lacking

    the Mlh1 protein were largely infertile. Since spermatogenesis

    arrested at metaphase I of meiosis and cells died by apoptosis,

    later germ cell stages were absent [48]. Nevertheless, spermato-

    genesis was still completed by a limited number of germ cells

    [49]. Although no studies of mutation in specific genes were per-

    formed, eggs fertilized with surviving sperm produced malformed

    embryos.

    Although fish that are homozygous mutant for the MMRgenesmlh1, msh2, and msh6 developed neoplasms particularly neurofi-

    bromas and malignant peripheral nerve sheath tumors of the eye

    and abdomen [50], mutagenesis induced by exogenous agents was

    not affected by loss of MMR gene function. For example, ethyl-

    nitrosourea (ENU) exposure is a standard protocol for generating

    germ line mutations in zebrafish. Although one might expect that

    successful mutagenesis would be enhanced by loss of MMR, defi-

    ciency in the MMRgene msh6 did not enhance ENU mutagenesis in

    thezebrafish germ line [51]. MMRalso contributed to growth arrest

    in response to DNAdamage by alkylating agents [52]. Although the

    primary response to alkylation damage occurs viaBER, invivo expo-

    sure of zebrafisheggs to alkylating agentsresulted in chromosomal

    instability and cell death due to stalled replication forks normally

    processed by MMR[53].

    2.5. Non homologous end joining

    NHEJ is a pathway that repairs double-strand breaks in DNAthat arise from exposure to UV, ionizing radiation (IR) or extreme

    damage from alkylating agents otherwise repaired by BER. This

    repair pathway is relatively inaccurate. The DNA ends are recog-

    nized by Ku70 and Ku80 that recruit the proteins that will perform

    end-rejoining without requiring a homologous template.ku80 and

    ku70 of the zebrafish NHEJ pathway have been cloned [54,55].

    Ku80 promoted survival of irradiated cells during embryogenesis.

    Radiation-induced apoptosis in the Ku80 knockdown zebrafish is

    suppressed by p53 knockdown, indicating that apoptosis result-

    ing from loss of Ku80 is p53 dependent [54]. Ku70 protein played

    a crucial role in protecting the developing nervous system from

    radiation-induced DNA damage during embryogenesis [54]. NHEJ

    is involved in retrotransposition of long interspersed elements

    (LINEs) from both zebrafish and mammalian sources. The use ofzebrafish LINEs enabled researchers to demonstrate the importance

    of Ku70 and Artemis in chicken B lymphocyte lines defective for

    these genes [56]. Furthermore, microinjection of linearized DNA

    encoding green fluorescent protein into zebrafish embryos was fol-

    lowed directly by fluorescence microscopy after which the DNA

    was recovered and sequenced. From these data Dai et al. concluded

    that NHEJ is the principal mechanism of exogenous gene integra-

    tion in zebrafish [57]. Recently, Liu et al., developed a visual-plus

    quantitative analysis systems to studyDNA DSB repairs in zebrafish,

    which also showed that NHEJ was predominant among the three

    DSB repair pathways (NHEJ,HR andSSA) in zebrafish embryos [58].

    NHEJ is the underlying mechanism for generating targeted

    mutations by new, highly efficient methods known as ZFNs

    (locus specific zing-finger nucleases) or TALENs (transcription

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    6/9

    Please cite this article in pressas: D.-S. Pei, P.R. Strauss, Zebrafish as a model system to study DNA damage and repair, Mutat. Res.: Fundam. Mol.

    Mech. Mutagen. (2012), http://dx.doi.org/10.1016/j.mrfmmm.2012.10.003

    ARTICLE IN PRESSGModel

    MUT-11229; No.of Pages9

    6 D.-S.Pei, P.R. Strauss / Mutation Researchxxx (2012) xxxxxx

    activator-like effector nucleases) in zebrafish and other eukary-

    otes [15]. The methodology is relatively simple, leads to indel

    (insertion/deletion) formation. Some of the genes that have been

    targeted with this methodology to date include kdr [14], golden

    and ntl [10,11], transferring receptor 2 (tfr2), dopamine trans-

    porter (slc6a3), telomerase (tert), hypoxia inducible factor 1a alpha

    (hif1aalpha), gridlock (hey2) [12,13], bmi1, Ikzf1, phf6(1), phf6(2),

    MyoD, andJak3 [15].

    2.6. Homologous recombination

    HR is the accurate, templated repair of DSBs in the late S-G2

    phase of the cell cycle. First a single-stranded region of DNA for

    strand invasion is generated, then a Holliday junction is formed

    and finally DNA synthesis is initiated using the intact strand as

    the template. HR has been reported in intact zebrafish embryos

    at early developmental stages [59] and in zebrafish ES cell cul-

    tures [60]. Several attempts to use HR to promote gene targeting in

    zebrafish have not met with success [61,62], although one recent

    report showed that knockdown of Rad51 decreased HR ability in

    zebrafish embryos [58]. Much remains tobe studied in thezebrafish

    HR system.

    2.7. Translesion synthesis

    The translesion polymerases enable the dividing cell to synthe-

    size past a mispair or a lesion during replication and/or repair. DNA

    polymerase eta was the first to be identified, as it is capable of

    bypassing thymine dimer CPDs. Unlike the replicative DNA poly-

    merases, , and , these polymerases exhibit low fidelity when

    copying undamaged DNA [6367]. Some like Rev1, however, insert

    a correct base (aC residue) opposite a template G but are inaccu-

    rate when synthesizing opposite an AP site or certain other minor

    groove adducts. DNA polymeraselambda andDNA polymerase mu,

    two additional translesion synthesis polymerases found in other

    vertebrates, have not yet been identified in the zebrafish genome.

    There is no published work on the translesion polymerases in

    zebrafishat this time.Giventhe rapidityof DNAsynthesisin thefirsthours after zebrafish fertilization, it seems likely that the transle-

    sion polymerases could play an important role [28].

    3. The role of BER pathway in zebrafish embryological

    development

    The role of the BER pathway in DNA repair (Fig. 1) is reviewed

    in the preceding section. In zebrafish unfertilized eggs and early

    stage embryos are able to perform at least the first three BER

    steps [26]. However, BERin early stage embryos (before 12hpf) has

    several unexpected features consistent with rapid cellular prolifer-

    ation (15min/cycle for the first 10 division cycles). In early stage

    embryos, aphidicolin-sensitive DNA polymerase(s) replace PolB,

    which is absent, while the presence of backup Mg2+-dependentendonuclease activity means that Apex1 has assistance in case of

    overwhelming oxidative damage. When the embryo hatches from

    the chorionic membrane and encounters normal oxidative stress,

    it switches to normal adult BER[27].

    Apex1 is a crucial participant in BER. The knockout of Apex1

    in mice is lethal, and no viable null cell line has been created,

    indicating that Apex1 is critical for development. Apex1 is highly

    conserved in different species and zebrafish. Zebrafish Apex1

    shares the same enzymatic properties as its human orthologue. In

    zebrafish,Apex1 protein has 78%homology(64% identity)with that

    of the human protein. Full knockdown of Apex1 in early zebrafish

    embryos leads to embryonic failure at the midblastula transition

    (MBT) stage. The MBT is the stage where cell division slows from

    15min/cycle, cells become motile, zygotic transcription is fully

    turned on, and spatial differentiation begins. Partial knockdown

    of Apex1 causes abnormalities in eyes, notochord, brain, blood

    cells and heart [26]. The failure of the heart to develop normally

    explains the basis for inability of mouse embryos to develop after

    Apex1 knockout: the heart forms early in development in mice and

    when heart development fails to proceed normally the embryo is

    resorbed efficiently. The advantage of the zebrafish system is that

    zebrafish embryos can continue developing for up to seven days

    with a non-functional heart. Both western blot and immunohisto-

    chemistry using anti-Apex1 antibody show that Apex1 is abundant

    in unfertilized eggs and throughout development (Fig. 2) [26].

    The use of zebrafish made it possible to discover that Apex1

    plays a critical and previously unknown role in cell physiology.

    These studies could not have been performedin mammals because

    of the developmental requirements described above. Apex1 reg-

    ulates the protein level of the crucial transcription factor Creb1

    [28]. Consequently, Apex1 regulates the levels of many other Creb-

    dependent proteins including PolB, the next protein in the BER

    pathway. After knockdown of Apex1, embryos fail to synthesize

    Creb1proteinand its binding partners,Creb1 binding protein (CBP),

    Creb1 modulating protein (Crem)and Targets of Creb1 (Torcs 1 and

    3).Similar effects areseen in primary cultures of mouse B cells from

    Apex1mice when remaining Apex1 is inhibited by CRT0044876.

    Finally, these results reinforce the requirement for endonuclease

    activity rather than any redox activity for successful embryogene-

    sis and cell survival because rescue requires microinjection of the

    gene for the endonuclease competent protein.

    4. Crosstalk between DNA repair and apoptosis

    Generation of reactive oxygen/nitrogen species and subsequent

    oxidativedamage to DNAare ongoing processesin every cell. These

    events are distinct from double strand breaks unless the breaks are

    very closely spaced or the damage is overwhelming. Nevertheless,

    oxidative damage requires repair to maintain genome integrity and

    stability. DNA damage activates checkpoint mechanisms to arrest

    the cell cycle in order to allow time to repair the DNA damage. Ifthe damageis toosevere forefficientrepair, thecell will be induced

    to undergo apoptosis, necrosis, autophagy, paraptosis, or possibly

    mitotic catastrophe. The tumor suppressor protein p53 provides

    one important signaling mechanism between DNA damage and

    apoptosis [72]. Loss or suppression of p53 or disabling mutations

    in p53 are associated with a variety of tumors [7378]. Despite

    the better understood importance for apoptosis, the p53 family

    serves broader and more complex functions than simply reducing

    theincidenceof cancer. It is involved in a numberof non-neoplastic

    processes including reproduction, metabolism, development, drug

    and radiation toxicity and aging [76,79,80].

    Zebrafish p53, which is structurally and functionally conserved

    with mammalian p53 [81], serves as a key mediator of apoptosis

    [76] especially for unrepaired double strand breaks. Most of thestudies involving p53 in zebrafish have involved a link to apopto-

    sis engendered b y either UV or IR radiation, both of which generate

    DSBs [76]. In that situationp53 mediates apoptosisthrough a linear

    pathway involving Bax transactivation, Bax translocation from the

    cytosol to membranes, cytochrome c release from mitochondria,

    and caspase-9 activation, followed by the activation of caspase-

    3, -6, and -7. p53-mediated apoptosis can be blocked at multiple

    checkpoints: by inhibiting p53 activity directly, by Bcl-2 family

    members regulating mitochondrial function, by E1B 19K blocking

    caspase-9 activation, and by caspase inhibitors [82].

    Regulation of p53 transcription is complex and not entirely

    understood [79]. Among the modulators in zebrafish are Mdm2

    [83] and 113p53, an alternative splicing p53 isoform [84].

    Instead of acting as a dominant negative for p53 [84],

    113p53

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    7/9

    Please cite this article in press as: D.-S. Pei, P.R. Strauss, Zebrafish as a model system to study DNA damage and repair, Mutat. Res.: Fundam. Mol.

    Mech. Mutagen. (2012), http://dx.doi.org/10.1016/j.mrfmmm.2012.10.003

    ARTICLE IN PRESSGModel

    MUT-11229; No.of Pages9

    D.-S. Pei,P.R. Strauss / Mutation Researchxxx (2012) xxxxxx 7

    Fig. 2. Analysis of theproteinexpressionlevel of Apex1 at different developmental stages in zebrafish by confocal microscopy. (A and B) Apex1, a maternal protein, is stored

    in the unfertilizedegg, as shown by fluorescence microscopy (A,FITC-labeled Apex1) and confocalmicroscopy (B, the merged picture withFITC-labeled Apex1 andpropidium

    iodide-stained nuclei); (C) 4-cell stage embryo with lateral view (up) and top view (lower right corner); (D) 64-cell stage with lateral view (up) and top view (lower right

    corner); (E and F) sphere stage (E) and 50% epiboly stage (F) with the animal pole to the top; (G and H) 10 somite stage (G) and 1 dpf stage (H) with dorsal to the top and

    anterior to the left; (I and J) confocal microscopy of a 64-cell stage embryo (J is theenlarged view of thesquare region in panel I.) The diploid nuclei areclearly visible with

    red color. (K and L) The 3.2kb upstream sequence preceding theATG start codon on the Apex promoter in zebrafishwas amplified and inserted into pEGFP-N3 to construct

    the pApexp3k-GFP plasmid. Constructs (2nl, 50ng/l) were micro-injected into 2cell stage embryos and GFPexpressionwas examined at 1dpf by.

    differentially modulated p53 target gene expression to antago-

    nize p53 apoptotic activity after zebrafish embryos were exposed

    to foreign DNA with broken ends or IR that generated DSBs

    [81,84]. Overexpression of113p53 enhanced p53-dependent

    up-regulation ofp21 and mdm2, but inhibited p53-dependent up-

    regulation of the proapoptotic gene bax [76,84].

    In zebrafisha common methodology forknocking downselected

    proteins in early embryogenesis is the microinjection of mor-

    pholino oligonucleotides (MO) designed to target a selected mRNA

    [6,85,86]. Unlike inhibitory RNA transfected into mammalian cells,

    theMO need notbe processedbut,instead, bindsdirectlyto itscom-plementary sequence to inhibit either splicing or translation. Since

    MO are stable and non-degradable and interact with target mRNA

    in a stoichiometric fashion, effects of knockdown can frequently be

    detected for up to seven dpf. As with any knockdown technology,

    nonspecific, offtarget effects occurso thatthe knockdown pheno-

    type needs to be confirmed by rescue with the appropriate, coding

    mRNA. Of interest here is that off target effects can frequently

    be corrected by knocking down p53 along with the gene of interest

    [87] or performing theknockdownin p53mutant (p53M214K/M214K )

    fish [88]. In this line, fish fail to undergo DNA damage-dependent

    apoptosis after -irradiation, fail to upregulate p21 after UV irra-diation and do not arrest at the G1/S checkpoint [89]. Given that

    microinjection of DNA with broken ends up-regulates p53 [81],

    off-target effects involving activation of p53 should not come as

    a surprise.

    Our recent study showed that loss of Apex1, one of major pro-

    teins in BER pathway, causes selective DNA damage and activates

    the p53 pathway (Pei&Strauss, unpublished data). Expression of

    p21, mdm2 and113p53 increased in Apex1 knock down embryos.

    The morphants had smaller heads and eyes and abnormal brains.

    The morphant phenotype was rescued with mRNA for human

    wild-type APEX1 [26]. More important, a similar morphant phe-

    notype was seen in p53 mutant (p53M214K/M214K ) fish and in wild

    type fish in which both p53 and Apex1 had been knocked down(Pei&Strauss, unpublished data). Clearly, Apex1 deficiency acti-

    vated a p53-dependent pathway but the brain and neural effects

    and the loss of Creb1 resulting from Apex1 loss were independent

    of p53-mediated apoptosis. On the basis of these results, we pro-

    pose that Apex1 not only regulates Creb1 but also that Apex1 be

    included as a regulator of p53.

    Inaddition, one mightalso ask whethertheendsof the mRNAin

    the double strand mRNA/MO complex might be digested away so

    that the resulting double strand fragment is recognized as a double

    strand break intermediate. The complex could act as a poison for

    the components of DSBR, since the morpholino-containing strand

    cannotbe degraded readily. Hence,it wouldtrigger a p53-mediated

    apoptotic response

    http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    8/9

    http://dx.doi.org/10.1007/s10695-10011-19598-10695http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1155/2012/407079http://localhost/var/www/apps/conversion/tmp/scratch_3/dx.doi.org/10.1016/j.mrfmmm.2012.10.003
  • 8/11/2019 Zebrafish as a Model System to Study DNA Damage and Repair

    9/9