We previously identified a Drosophila maternal effect-lethal mutant named ‘no poles’ (nopo). Embryos from nopo females undergo mitotic arrest with barrel-shaped, acentrosomal spindles during the rapid cycles of syncytial embryogenesis because of activation of a Chk2-mediated DNA checkpoint. NOPO is the Drosophila homolog of human TNF receptor associated factor (TRAF)-interacting protein (TRIP), which has been implicated in TNF signaling. NOPO and TRIP contain RING domains closely resembling those of known E3 ubiquitin ligases. We herein sought to elucidate the mechanism by which TRIP/NOPO promotes genomic stability by performing a yeast two-hybrid screen to identify potential substrates/interactors. We identified members of the Y-family of DNA polymerases that facilitate replicative bypass of damaged DNA (translesion synthesis) as TRIP interactors. We show that TRIP and NOPO co-immunoprecipitate with human and Drosophila Polη, respectively, from cultured cells. We generated a null mutation in Drosophila Polη (dPolη) and found that dPolη-derived embryos have increased sensitivity to ultraviolet irradiation and exhibit nopo-like mitotic spindle defects. dPolη and nopo interact genetically in that overexpression of dPolη in hypomorphic nopo-derived embryos suppresses nopo phenotypes. We observed enhanced ubiquitylation of Polη by TRIP and NOPO E3 ligases in human cells and Drosophila embryos, respectively, and show that TRIP promotes hPolη localization to nuclear foci in human cells. We present a model in which TRIP/NOPO ubiquitylates Polη to positively regulate its activity in translesion synthesis.

The early embryonic development of organisms such as Drosophila melanogaster, Xenopus laevis, Danio rerio and Caenorhabditis elegans are characterized by rapid progression through the cell cycle (Budirahardja and Gönczy, 2009; O’Farrell et al., 2004). In Drosophila, early embryonic cell cycles consist of 13 oscillating rounds of DNA replication and mitosis without intervening gap phases or cytokinesis. Cell-cycle regulators are provided maternally and are slowly depleted until the midblastula transition. meiotic 41 (mei-41) and grapes (grp), which encode Drosophila homologs of ATM and Rad3-related (ATR) and Checkpoint 1 (Chk1) kinases, respectively, are essential effectors of a DNA damage/replication checkpoint pathway required to slow late syncytial cell cycles (11-13) in order to introduce a G2 phase (Fogarty et al., 1994; Sibon et al., 1999; Sibon et al., 1997).

The capacity of cells to respond to potentially detrimental DNA damage is of crucial importance for efficient and accurate replication of the genetic material. Most eukaryotic cells employ highly regulated responses to damaged DNA involving ATR and ataxia telangiectasia mutated (ATM), and their effector kinases Chk1 and Checkpoint 2 (Chk2), which trigger signaling cascades to respond to and repair damage (Abraham, 2001; Sancar et al., 2004). During the rapid divisions of syncytial embryogenesis, however, there may be insufficient time for canonical checkpoints to facilitate DNA repair (O’Farrell et al., 2004). Because accumulated DNA damage may block progression of replication forks, cells possess mechanisms to temporarily tolerate DNA damage until it can be repaired (Sale et al., 2012; Waters et al., 2009).

Translesion synthesis (TLS), which promotes completion of DNA replication when the replication fork encounters damage, is one mechanism used by the cell to bypass damage during S phase (Sale et al., 2012; Waters et al., 2009). This process is initiated by proliferating cell nuclear antigen (PCNA) mono-ubiquitylation, which triggers recruitment of specialized DNA polymerases to the replication fork (Freudenthal et al., 2010; Kannouche et al., 2004). These polymerases include Polξ (also referred to as Rev3), a member of the B-family of DNA polymerases, and members of the Y-family of DNA polymerases: Rev1, Polι, Polη and Polκ (Kannouche and Lehmann, 2006; Lehmann et al., 2007; Sale et al., 2012; Shaheen et al., 2010). Mutation of human Polη results in a variant form of xeroderma pigmentosum (XP-V), a disease characterized by increased UV sensitivity and susceptibility to skin cancers (Kannouche et al., 2001; Kannouche and Stary, 2003; Masutani et al., 1999).

Three members of the Y-family of DNA polymerases are conserved in Drosophila: dPolη, dPolι and dRev1 (Ishikawa et al., 2001). dPolη and dPolι are functional translesion polymerases in vitro (Ishikawa et al., 2001). dPolη larvae exhibit increased sensitivity to UV irradiation, and homologous recombination is impaired in dPolη flies (Kane et al., 2012). Germline knockdown of polh-1, the C. elegans Polη homolog, causes increased sensitivity of early embryos to UV irradiation (Ohkumo et al., 2006). Recent studies suggest that POLH-1 functions in C. elegans to keep the rapid cell cycles of early embryogenesis on schedule (Holway et al., 2006; Kim and Michael, 2008).

We previously described a Drosophila maternal effect-lethal mutant named ‘no poles’ (nopo) (Merkle et al., 2009). Embryos from nopo females undergo mitotic arrest during the rapid S-M cycles of syncytial embryogenesis. Chk2 plays a checkpoint function in the Drosophila early embryo with localized activation in response to DNA damage or incomplete replication (Sibon et al., 2000; Takada et al., 2003). We found evidence of Chk2 activation in nopo-derived embryos, suggesting that NOPO promotes genomic maintenance in early embryos (Merkle et al., 2009). Based on the abnormally short interphase 11 of nopo-derived embryos, we hypothesized that they enter mitosis before completing DNA replication, thereby activating Chk2.

nopo, which encodes a RING domain-containing candidate E3 ubiquitin ligase, is the Drosophila homolog of the human gene encoding TRAF-interacting protein (TRIP). Tumor necrosis factor receptor (TNFR)-associated factors (TRAFs) are key adaptor molecules in the TNF-signaling pathway that promote cell proliferation, activation, differentiation, and apoptosis (Ha et al., 2009). Whether TRIP plays a role in TNF signaling, however, is unclear (Lee et al., 1997; Regamey et al., 2003).

Although TRIP/NOPO substrates have not been reported to date, mouse TRIP is a functional E3 ligase in vitro (Besse et al., 2007). E3 ligases facilitate transfer of ubiquitin from an E2 conjugating enzyme to substrates (Fang and Weissman, 2004). Ubiquitylation plays important roles in many cellular processes, including protein processing, cell-cycle control, chromatin remodeling, DNA repair and membrane trafficking by targeting proteins for destruction by the 26S proteasome, altering subcellular localizations, and/or changing protein-protein interactions (Acconcia et al., 2009; Al-Hakim et al., 2010; Broemer and Meier, 2009; Glickman and Ciechanover, 2002; Hershko, 1997; Le Bras et al., 2011; O’Connell and Harper, 2007).

To elucidate the mechanism by which the TRIP/NOPO E3 ligases promote genomic stability, we performed a yeast two-hybrid screen using human TRIP as bait. We report herein the interaction of human TRIP and Drosophila NOPO with members of the Y-family of DNA polymerases in yeast two-hybrid assays and their co-immunoprecipitation from cultured cells. We generated a null allele of Drosophila Polη (dPolη) and found that dPolη-derived embryos are sensitive to UV irradiation and have nopo-like mitotic spindle defects. We find that overexpression of dPolη in early embryos compensates for partial loss of NOPO. We show that TRIP/NOPO enhances ubiquitylation of human Polη (hPolη) and dPolη in cultured cells and Drosophila embryos, respectively, and that TRIP promotes localization of hPolη into nuclear foci in cultured human cells. These findings suggest that TRIP/NOPO plays an important role in positively regulating the DNA damage tolerance activity of Polη during Drosophila embryogenesis and in human cells.

Drosophila NOPO and human TRIP are functional homologs

Embryos derived from Drosophila nopo females fail to progress to larval stages due to mitotic arrest with barrel-shaped, acentrosomal spindles during the rapid S-M cycles of syncytial embryogenesis (Merkle et al., 2009). We previously demonstrated rescue of nopo mutants by using a transgenic line carrying nopo genomic sequence (Merkle et al., 2009). Herein we generated a new transgenic line in which nopo cDNA was placed under control of the endogenous nopo promoter (Fig. 1A). Embryonic expression of this transgene was confirmed by RT-PCR (Fig. 1B). Embryos produced by nopo females were rescued by the presence of the nopo cDNA transgene, exhibiting a 76% hatch rate (embryo-to-larva transition) compared with 0% and 94% for nopo and wild-type females, respectively (Fig. 1C).

Fig. 1.

Transgenic human TRIP expression rescues Drosophila nopo mutants. (A) Schematic of the nopo gene region (top). The black boxes represent coding regions, white boxes represent 5′ and 3′ untranslated regions (UTRs), and lines represent introns. Transgenic rescue constructs (bottom) contain genomic flanking sequence (lines), 5′- and 3′-UTRs of nopo (white boxes), and nopo or TRIP open reading frame (ORF; dashed lines). (B) RT-PCR confirming nopo or TRIP transgenic expression in nopoExc142-derived embryos (0-2 hours). Positive and negative controls: wild-type and nopoExc142-derived embryos, respectively. Loading control: RP49. (C) nopo or TRIP expression restored fertility to nopoExc142 females. (D) nopo or TRIP expression rescued aberrant spindles of nopoExc142-derived embryos. (E) Embryos stained for Centrosomin (blue), α-tubulin (green) and DNA (red). Representative mitotic spindles in embryos from females of indicated genotypes are shown. Asterisks, P<0.0001 (indicated pairwise comparisons). Scale bar: 10 μm. wt, wild type.

Fig. 1.

Transgenic human TRIP expression rescues Drosophila nopo mutants. (A) Schematic of the nopo gene region (top). The black boxes represent coding regions, white boxes represent 5′ and 3′ untranslated regions (UTRs), and lines represent introns. Transgenic rescue constructs (bottom) contain genomic flanking sequence (lines), 5′- and 3′-UTRs of nopo (white boxes), and nopo or TRIP open reading frame (ORF; dashed lines). (B) RT-PCR confirming nopo or TRIP transgenic expression in nopoExc142-derived embryos (0-2 hours). Positive and negative controls: wild-type and nopoExc142-derived embryos, respectively. Loading control: RP49. (C) nopo or TRIP expression restored fertility to nopoExc142 females. (D) nopo or TRIP expression rescued aberrant spindles of nopoExc142-derived embryos. (E) Embryos stained for Centrosomin (blue), α-tubulin (green) and DNA (red). Representative mitotic spindles in embryos from females of indicated genotypes are shown. Asterisks, P<0.0001 (indicated pairwise comparisons). Scale bar: 10 μm. wt, wild type.

To test for functional conservation of human TRIP and Drosophila NOPO E3 ubiquitin ligases, we investigated whether transgenic expression of human TRIP under control of the endogenous nopo promoter could rescue Drosophila nopo mutants (Fig. 1A). Embryonic expression of this transgene was confirmed by RT-PCR (Fig. 1B). Fertility was restored to nopo females carrying the TRIP cDNA transgene, resulting in an 83% hatch rate (Fig. 1C). Mitotic spindles were also restored to a wild-type morphology: whereas 94% of embryos from nopo females exhibited abnormal spindle morphology, the frequency was reduced to 22% and 14% in embryos carrying the nopo or TRIP transgenes, respectively (Fig. 1D,E). These data suggest that human TRIP is a functionally conserved homolog of Drosophila NOPO.

TRIP interacts with Y-family DNA polymerases

RING domain-containing E3 ligases bind directly to their substrates to catalyze direct transfer of ubiquitin from an E2 ubiquitin conjugating enzyme (Deshaies and Joazeiro, 2009). To elucidate the role of NOPO in maintaining genomic integrity during early embryonic development in Drosophila, we sought to identify potential substrate(s) of the NOPO E3 ubiquitin ligase by performing a yeast two-hybrid screen. Due to a lack of commercially available, high-quality cDNA libraries prepared from Drosophila early embryos, we chose to carry out our yeast-two hybrid screen by using TRIP, the human homolog of NOPO, as the bait and a HeLa cell cDNA library as the prey. Sequence analysis revealed that one of the first colonies to appear on selective media in this screen contained a clone encoding a C-terminal fragment of hPolκ, a member of the Y-family of DNA polymerases.

To confirm the interaction of TRIP with hPolκ and to determine if TRIP interacts with other members of the Y-family of DNA polymerases (hPolη, hPolι and hRev1), we performed yeast two-hybrid assays using TRIP as bait with full-length or truncated versions of these polymerases as prey (illustrated for hPolη in Fig. 2A). We found that TRIP interacted with the C-terminal half of hPolκ, but not with the N-terminal half or full-length form of this polymerase (Fig. 2B). Similarly, TRIP interacted with the C-terminal half of hPolη; we also observed interaction between TRIP and the full-length form, but not the N-terminal half, of hPolη (Fig. 2C). These data are consistent with previous reports demonstrating the existence of several protein-protein interaction domains in the C-terminal regions of Y-family DNA polymerases (Waters et al., 2009). Our results for hPolι and hRev1 were inconclusive (data not shown).

Fig. 2.

TRIP interacts with human Y-family DNA polymerases. (A) Schematic representation of full-length and truncated forms of hPolη (prey). (B,C) Yeast two-hybrid assays. Yeast expressing human TRIP (BD, ‘bait’) and full-length or truncated forms of hPolκ (B) or hPolη (C) (AD, ‘prey’) were spotted onto selective triple dropout (TDO) media to assess interactions. Plating on double dropout (DDO) media confirmed presence of bait and prey constructs. Negative control: SV40 T-antigen. (D,E) hPolη and TRIP co-immunoprecipitation. (D) HeLa cells were co-transfected with indicated plasmids. TRIP-MYC immunoprecipitates and whole cell lysates were analyzed by immunoblotting. (E) Reciprocal co-IP of FLAG-hPolη complexes from HeLa cells co-transfected as indicated. IB, immunoblotting; IP, immunoprecipitates; WCL, whole cell lysates.

Fig. 2.

TRIP interacts with human Y-family DNA polymerases. (A) Schematic representation of full-length and truncated forms of hPolη (prey). (B,C) Yeast two-hybrid assays. Yeast expressing human TRIP (BD, ‘bait’) and full-length or truncated forms of hPolκ (B) or hPolη (C) (AD, ‘prey’) were spotted onto selective triple dropout (TDO) media to assess interactions. Plating on double dropout (DDO) media confirmed presence of bait and prey constructs. Negative control: SV40 T-antigen. (D,E) hPolη and TRIP co-immunoprecipitation. (D) HeLa cells were co-transfected with indicated plasmids. TRIP-MYC immunoprecipitates and whole cell lysates were analyzed by immunoblotting. (E) Reciprocal co-IP of FLAG-hPolη complexes from HeLa cells co-transfected as indicated. IB, immunoblotting; IP, immunoprecipitates; WCL, whole cell lysates.

To further verify these interactions, we co-expressed MYC-tagged TRIP and epitope-tagged hPolη and hPolκ (HA- or FLAG-tagged) in HeLa cells for co-immunoprecipitation experiments. We detected the presence of HA-hPolη in TRIP-MYC immunoprecipitates; reciprocally, we detected the presence of TRIP-MYC in FLAG-hPolη immunoprecipitates, thereby confirming the yeast two-hybrid interaction between TRIP and hPolη in this system (Fig. 2E,F). We were unable, however, to detect co-immunoprecipitation of tagged hPolκ and MYC-TRIP (data not shown). We frequently observed a faint, more slowly migrating form of tagged hPolη on immunoblots of cell lysates; this band may represent a mono-ubiquitylated form of hPolη as reported previously (Bienko et al., 2010).

NOPO interacts with Y-family DNA polymerases

Three of the four members of the human Y-family DNA polymerases are conserved in Drosophila: Polη, Polι and Rev1 (Fig. 3A). To determine if NOPO, like its human homolog, interacts with the Drosophila Y-family DNA polymerases, we performed yeast two-hybrid assays using NOPO as bait with full-length or truncated versions of each of these polymerases as prey (as illustrated for dPolη in Fig. 3B). We found that NOPO interacted with the C-terminal halves and full-length forms of both dPolη and dPolι; results for dRev1 were inconclusive (Fig. 3C,D; data not shown). To further verify these interactions, we co-expressed MYC-tagged NOPO and HA-tagged dPolη or dPolι in Drosophila S2 cells. We detected the presence of HA-dPolη, but not HA-dPolι, in NOPO-MYC immunoprecipitates, thereby confirming the yeast two-hybrid interaction between NOPO and dPolη in this system (Fig. 3E; data not shown).

Fig. 3.

NOPO interacts with Drosophila Y-family DNA polymerases. (A) Conservation of human and Drosophila Y-family DNA polymerases. (B) Schematic representation of full-length and truncated forms of dPolη (prey). (C,D) Yeast two-hybrid assays. Yeast expressing NOPO (BD, ‘bait’) and full-length or truncated forms of dPolη (C) or dPolι (D) (AD, ‘prey’) were spotted onto selective triple dropout (TDO) media to assess interactions. Plating on double dropout (DDO) media confirmed presence of bait and prey constructs. Negative control: SV40 T-antigen. (E) dPolη and NOPO co-immunoprecipitation. S2 cells were co-transfected as indicated. NOPO-MYC immunoprecipitates and whole cell lysates were resolved by immunoblotting.

Fig. 3.

NOPO interacts with Drosophila Y-family DNA polymerases. (A) Conservation of human and Drosophila Y-family DNA polymerases. (B) Schematic representation of full-length and truncated forms of dPolη (prey). (C,D) Yeast two-hybrid assays. Yeast expressing NOPO (BD, ‘bait’) and full-length or truncated forms of dPolη (C) or dPolι (D) (AD, ‘prey’) were spotted onto selective triple dropout (TDO) media to assess interactions. Plating on double dropout (DDO) media confirmed presence of bait and prey constructs. Negative control: SV40 T-antigen. (E) dPolη and NOPO co-immunoprecipitation. S2 cells were co-transfected as indicated. NOPO-MYC immunoprecipitates and whole cell lysates were resolved by immunoblotting.

Drosophila Y-family polymerases are expressed in early embryos

The yeast two-hybrid interactions and co-immunoprecipitations observed between TRIP/NOPO E3 ubiquitin ligases and Y-family DNA polymerases suggested that these proteins might function in a common pathway to preserve genomic integrity. To determine if these polymerases are expressed during Drosophila early embryogenesis, we performed RT-PCR using mRNA extracted from 0-2 hour embryos. We found that, like nopo, all of the Y-family DNA polymerases are expressed during syncytial embryogenesis (Fig. 4A).

Fig. 4.

Drosophila Y-family DNA polymerase expression and localization. (A) RT-PCR analysis of dPolη, dPolι and dRev1 in embryos (0-2 hour). Positive controls: RP49 and nopo. (B) dPolη developmental expression. Mean relative expression (normalized against RP49) ± s.e.m. is displayed. (C) Embryos from females carrying UASp-HA-dPolη and nos-Gal4 immunostained for HA (green) and DNA (magenta). HA-dPolη localized to interphase nuclei (top panels) but was undetectable during mitosis (bottom panels). Scale bar: 10 μm.

Fig. 4.

Drosophila Y-family DNA polymerase expression and localization. (A) RT-PCR analysis of dPolη, dPolι and dRev1 in embryos (0-2 hour). Positive controls: RP49 and nopo. (B) dPolη developmental expression. Mean relative expression (normalized against RP49) ± s.e.m. is displayed. (C) Embryos from females carrying UASp-HA-dPolη and nos-Gal4 immunostained for HA (green) and DNA (magenta). HA-dPolη localized to interphase nuclei (top panels) but was undetectable during mitosis (bottom panels). Scale bar: 10 μm.

Mutations in hPolη result in a variant form of xeroderma pigmentosum, a disease characterized by an increased incidence of UV-induced skin cancer. Due to the key role of Polη in TLS and its link to human disease, as well as the lack of available mutants to study the functions of the other Drosophila Y-family polymerases, we chose to focus the remainder of our study on dPolη. To determine the tissues and developmental stages in which dPolη is expressed, we performed RT-PCR using mRNA isolated from embryos of various stages, larvae, pupae, ovaries, testes, and male and female carcasses. We found that dPolη is most highly expressed in 0-2 hour embryos and ovaries, suggesting that it could potentially interact with NOPO in vivo to promote maintenance of genomic integrity during early embryogenesis (Fig. 4B).

To determine the localization of dPolη in early embryos, we generated a transgenic line for expression of HA-tagged full-length dPolη under control of the UASp promoter (Rørth, 1998). Using nanos-Gal4:VP16 (‘nos-Gal4’) for germline expression of this transgene, we performed immunostaining of syncytial embryos and found HA-dPolη to localize within interphase nuclei; no signal was detected on mitotic chromosomes (Fig. 4C). These observations are consistent with the previously reported localization of hPolη in cultured cells (Kannouche et al., 2003; Kannouche and Lehmann, 2006).

dPolη-derived embryos are sensitive to DNA-damaging agents and have nopo-like defects

To determine if dPolη, like nopo, is required during early embryogenesis, we generated a null allele of dPolη (Exc2.15) via imprecise excision of P-element EY07711 in the 5′ UTR of the dPolη gene (Fig. 5A; supplementary material Fig. S1A). Homozygous dPolηExc2.15 females are viable and fertile, producing embryos with wild-type hatch rates; similar results were obtained for females carrying dPolηExc2.15 in trans to Df(3L)BSC284, which uncovers dPolη (supplementary material Fig. S1B). This observation is consistent with studies of POLH-1 during C. elegans embryogenesis and suggests that functional redundancy among Y-family polymerase members may account for the lack of severe phenotypes in single mutants (Roerink et al., 2012).

Fig. 5.

Increased UV sensitivity and nopo-like spindles of dPolη mutants. (A) Schematic of dPolη gene region. The black boxes represent coding regions, white boxes represent 5′ and 3′ untranslated regions (UTRs), and lines represent introns. dPolηExc2.15 (gap represents deleted region) was generated by imprecise excision of P-element EY07711 (triangle). (B) UV sensitivity assay. Hatch rates of UV-irradiated embryos were normalized to untreated controls for each genotype; hatch rates of untreated controls were ∼95% (supplementary material Fig. S1; data not shown). Mean values from ≥3 independent experiments are displayed. Error bars, s.d. (C) Quantification of mitotic index in untreated or UV-irradiated embryos. Asterisks, P<0.0001 (relative to untreated controls). (D-G) Representative mitotic spindles in embryos stained for α-tubulin (green) and DNA (red). Arrows mark detached centrosomes. (H) Quantification of nopo-like spindles in untreated or UV-irradiated embryos. Data are plotted as percentage of embryos with >50% nopo-like spindles. Phenotypes presented in C-H were observed throughout syncytial embryogenesis. For bar graphs: asterisks, P<0.0001 (compared to untreated or UV-irradiated wild-type control). Df=Df(3L)BSC284. Scale bar: 10 μm.

Fig. 5.

Increased UV sensitivity and nopo-like spindles of dPolη mutants. (A) Schematic of dPolη gene region. The black boxes represent coding regions, white boxes represent 5′ and 3′ untranslated regions (UTRs), and lines represent introns. dPolηExc2.15 (gap represents deleted region) was generated by imprecise excision of P-element EY07711 (triangle). (B) UV sensitivity assay. Hatch rates of UV-irradiated embryos were normalized to untreated controls for each genotype; hatch rates of untreated controls were ∼95% (supplementary material Fig. S1; data not shown). Mean values from ≥3 independent experiments are displayed. Error bars, s.d. (C) Quantification of mitotic index in untreated or UV-irradiated embryos. Asterisks, P<0.0001 (relative to untreated controls). (D-G) Representative mitotic spindles in embryos stained for α-tubulin (green) and DNA (red). Arrows mark detached centrosomes. (H) Quantification of nopo-like spindles in untreated or UV-irradiated embryos. Data are plotted as percentage of embryos with >50% nopo-like spindles. Phenotypes presented in C-H were observed throughout syncytial embryogenesis. For bar graphs: asterisks, P<0.0001 (compared to untreated or UV-irradiated wild-type control). Df=Df(3L)BSC284. Scale bar: 10 μm.

Polη is known to specifically bypass UV-induced lesions in yeast and mammals, and dPolη has been shown to synthesize efficiently past cyclobutane pyrimidine dimers in vitro (Ishikawa et al., 2001; Johnson et al., 1999; McCulloch et al., 2004; McDonald et al., 1997). We therefore tested the sensitivity of embryos from dPolηExc2.15 females to various doses of UV irradiation. Hatch rates of embryos from both wild-type and dPolηExc2.15 females decreased following UV exposure, but dPolηExc2.15-derived embryos were significantly more affected; germline expression of HA-dPolη restored a wild-type degree of UV sensitivity to dPolηExc2.15-derived embryos (Fig. 5B). Immunostaining revealed a significant increase in mitotic index in response to UV irradiation in dPolηExc2.15-derived embryos that was not seen in wild-type embryos; this effect was also rescued by germline expression of HA-dPolη (Fig. 5C). These findings are consistent with previous work showing that a deletion in dPolη caused increased sensitivity of third-instar larvae to UV irradiation (Kane et al., 2012).

In contrast to nopo-derived embryos, unperturbed dPolηExc2.15-derived embryos appeared to progress through syncytial embryogenesis at a normal rate and hatched normally (supplementary material Fig. S1B; Fig. S2). Immunostaining, however, revealed that a majority of spindles in 22% of untreated dPolηExc2.15-derived embryos (compared with 5% of wild-type embryos) had nopo-like defects (Fig. 5D-H). Upon exposure to 250 J/m2 UV, this frequency increased to 90% for dPolηExc2.15-derived embryos compared with 24% for wild-type embryos (Fig. 2H). Germline expression of HA-dPolη restored the frequency of spindle defects to wild-type levels in dPolηExc2.15-derived embryos with or without UV treatment (Fig. 5H).

We previously reported that syncytial embryos from nopo females undergo Chk2-mediated mitotic arrest (Merkle et al., 2009). Chk2 is activated in Drosophila syncytial embryos in response to mitotic entry with incompletely replicated or damaged DNA (Sibon et al., 2000; Takada et al., 2003). We showed that nopo defects could be suppressed by mutation of maternal nuclear kinase (mnk; lok - FlyBase), which encodes the Drosophila Chk2 homolog, suggesting that NOPO promotes genomic integrity during early embryogenesis (Abdu et al., 2002; Brodsky et al., 2004; Masrouha et al., 2003; Merkle et al., 2009; Xu et al., 2001). To determine whether Chk2 mediates the mitotic defects of dPolηExc2.15-derived embryos, we generated flies mutant for both dPolη and mnk. The mild nopo-like spindle phenotype observed in unchallenged dPolηExc2.15-derived embryos was restored to wild-type levels in embryos from double-mutant females; mnk also suppressed the spindle abnormalities in UV-irradiated embryos from dPolηExc2.15 females (Fig. 5H). These data suggest that dPolη is required for normal progression through mitosis when DNA damage is present during early embryogenesis.

Human Polη is recruited to replication forks and is crucial for S-phase progression in response to hydroxyurea (HU)-induced DNA replication stress (Kane et al., 2012). We therefore tested the sensitivity of dPolηExc2.15 larvae to HU. We found that, similar to mei-41RT1 control larvae, dPolηExc2.15 larvae were sensitive to HU treatment, exhibiting a significant reduction in survival compared with expected Mendelian ratios (supplementary material Fig. S3). These results suggest that dPolη plays an important role in responding to HU-induced DNA damage.

dPolη overexpression suppresses nopo phenotypes

To further explore the possibility of a functional relationship between nopo and dPolη, we looked for genetic interactions following germline overexpression of HA-dPolη in females homozygous for nopoSZ3004, a hypomorphic allele (Merkle et al., 2009). Embryos from nopoSZ3004 females carrying either nos-Gal4 or UASp-HA-dPolη transgenes exhibited decreased hatch rates of 42% and 44%, respectively, compared with the wild-type hatch rate of 94% (Fig. 6A). Mitotic spindle defects occurred at a frequency of 43% and 48% in embryos from nopoSZ3004 females carrying either nos-Gal4 or UASp-HA-dPolη transgenes, respectively, compared with a wild-type frequency of 15% (Fig. 6B). Both phenotypes were strongly suppressed in embryos from nopoSZ3004 females with germline overexpression of HA-dPolη: hatch rates increased to 90%, and the frequency of mitotic spindle defects decreased to 25% (Fig. 6A,B).

Fig. 6.

Suppression of nopo by dPolη overexpression. (A) germline dPolη overexpression restored fertility to nopoSZ3004 females. (B,C) Embryos (B, 0-2 hour; C, 3-5 hour) were stained for α-tubulin and DNA. Mitotic spindle abnormalities (B) and early developmental arrest (C) of nopoSZ3004-derived embryos were rescued by germline dPolη overexpression. Single asterisks, P<0.001; double asterisks, P<0.0001 (compared to wild type).

Fig. 6.

Suppression of nopo by dPolη overexpression. (A) germline dPolη overexpression restored fertility to nopoSZ3004 females. (B,C) Embryos (B, 0-2 hour; C, 3-5 hour) were stained for α-tubulin and DNA. Mitotic spindle abnormalities (B) and early developmental arrest (C) of nopoSZ3004-derived embryos were rescued by germline dPolη overexpression. Single asterisks, P<0.001; double asterisks, P<0.0001 (compared to wild type).

Embryos from nopoSZ3004 females carrying nos-Gal4 or UASp-HA-dPolη transgenes were developmentally delayed with only 13% or 15%, respectively, reaching gastrulation by 5 hours after egg deposition compared with 93% of wild-type embryos (Fig. 6C). Germline overexpression of HA-dPolη in nopoSZ3004 females suppressed this developmental delay with 84% of embryos reaching gastrulation by 5 hours after egg deposition (Fig. 6C). Taken together, these data suggest that dPolη overexpression can compensate for the loss of nopo and that dPolη and NOPO contribute to the same biological process. Conversely, we did not observe enhancement of nopoSZ3004 phenotypes in nopoSZ3004 dPolη double mutants (data not shown).

TRIP/NOPO E3 ubiquitin ligase enhances Polη ubiquitylation

We considered the possibility that Polη might be a substrate of TRIP/NOPO E3 ubiquitin ligase. Using a cell-based, polyhistidine-ubiquitin assay, we tested whether TRIP overexpression in cultured human cells could promote Polη ubiquitylation (Campanero and Flemington, 1997; Treier et al., 1994). Without TRIP overexpression, we detected a single ubiquitylated species of hPolη, consistent with previous studies demonstrating its mono-ubiquitylation (Fig. 7A) (Bienko et al., 2010). TRIP overexpression caused further ubiquitylation of HA-hPolη as evidenced by its upward laddering and increased signal intensity in this assay. These data suggest that TRIP promotes hPolη ubiquitylation.

Fig. 7.

TRIP/NOPO enhances Polη ubiquitylation. (A) His-ubiquitin assay in HeLa cells. Cells co-transfected as indicated were lysed under denaturing conditions. Ubiquitylated proteins recovered by nickel affinity chromatography (Ni-NTA) and whole cell lysates were analyzed by immunoblotting. (B) His-ubiquitin assay in Drosophila embryos. Assays were performed as in A using embryos from wild-type or nopoExc142 females with germline expression of HA-dPolη ± His7-ubiquitin (His7-Ub). IB, immunoblotting; WCL, whole cell lysates.

Fig. 7.

TRIP/NOPO enhances Polη ubiquitylation. (A) His-ubiquitin assay in HeLa cells. Cells co-transfected as indicated were lysed under denaturing conditions. Ubiquitylated proteins recovered by nickel affinity chromatography (Ni-NTA) and whole cell lysates were analyzed by immunoblotting. (B) His-ubiquitin assay in Drosophila embryos. Assays were performed as in A using embryos from wild-type or nopoExc142 females with germline expression of HA-dPolη ± His7-ubiquitin (His7-Ub). IB, immunoblotting; WCL, whole cell lysates.

Ubiquitylation of Y-family DNA polymerases has been shown in budding yeast, C. elegans and mammalian cells, but not in Drosophila (Skoneczna et al., 2007; Kim and Michael, 2008; Jung et al., 2010; Bienko et al., 2010). To determine whether dPolη is ubiquitylated during Drosophila embryogenesis, we used an in vivo, polyhistidine-ubiquitin assay. We found that HA-dPolη was ubiquitylated in syncytial embryos from wild-type females with germline expression of HA-dPolη and His7-Ub; by contrast, nopoExc142-derived embryos showed a significant, albeit modest, decrease in the level of ubiquitylated dPolη in this assay (Fig. 7B; supplementary material Fig. S4). These data suggest that NOPO promotes ubiquitylation of dPolη during early embryogenesis. Because nopoExc142-derived embryos arrest during syncytial embryogenesis, however, we cannot rule out the possibility that observed changes in dPolη ubiquitylation in nopoExc142-derived embryos might be a secondary effect.

hPolη colocalizes with TRIP in cultured human cells

We previously reported that N-terminally tagged eGFP-TRIP localizes to nuclear puncta during G2 phase in transfected HeLa cells (Merkle et al., 2009). Another group reported that endogenous TRIP localizes to nucleoli in MCF7(BD) breast epithelial cells (Zhou and Geahlen, 2009). We confirmed their results by expressing C-terminally tagged TRIP-mCherry in HeLa cells and colocalizing TRIP with a nucleolar marker (Fig. 8A). We occasionally observed localization of TRIP-mCherry to nuclear puncta (Fig. 8C).

Fig. 8.

TRIP promotes recruitment of hPolη to nuclear foci. (A,B) HeLa cells expressing TRIP-mCherry (A) or GFP-hPOLη (B) (green) stained for fibrillarin (red; nucleolar marker) and DNA (blue) display enriched nucleolar localization of both fusion proteins. (C) HeLa cells co-expressing GFP-hPOLη (green) and TRIP-mCherry (blue) display colocalization of GFP-hPOLη with PCNA foci. (D) Increased frequency of GFP-hPOLη foci upon TRIP-mCherry co-expression. Asterisk, P<0.001. (E) Similar frequency of endogenous PCNA foci in cells expressing GFP-hPOLη ± TRIP-mCherry. NS, nonsignificant.

Fig. 8.

TRIP promotes recruitment of hPolη to nuclear foci. (A,B) HeLa cells expressing TRIP-mCherry (A) or GFP-hPOLη (B) (green) stained for fibrillarin (red; nucleolar marker) and DNA (blue) display enriched nucleolar localization of both fusion proteins. (C) HeLa cells co-expressing GFP-hPOLη (green) and TRIP-mCherry (blue) display colocalization of GFP-hPOLη with PCNA foci. (D) Increased frequency of GFP-hPOLη foci upon TRIP-mCherry co-expression. Asterisk, P<0.001. (E) Similar frequency of endogenous PCNA foci in cells expressing GFP-hPOLη ± TRIP-mCherry. NS, nonsignificant.

Due to its low expression levels, hPolη is difficult to detect by immunofluorescence; therefore, its localization is typically assessed in transfected cells overexpressing hPolη (Bienko et al., 2010; Kannouche et al., 2002; Lehmann et al., 2007). To confirm that eGFP-hPolη overexpression does not induce DNA damage, we assessed the presence of γ-H2AX foci, which appear in response to double-stranded breaks. We found that eGFP-hPolη-expressing cells exhibited a decreased (rather than increased) frequency of γ-H2AX-foci (supplementary material Fig. S5); this observation is consistent with a previously described role for hPolη in maintaining genomic stability during unperturbed S phase (Rey et al., 2009).

In cells expressing eGFP-hPolη, hPolη exhibited a diffuse nuclear localization with nucleolar enrichment (Fig. 8B). We occasionally observed hPolη localization to nuclear foci, consistent with its recruitment to stalled replication forks, where it interacts with PCNA during translesion synthesis (data not shown) (Kannouche et al., 2003; Kannouche and Lehmann, 2006). Indeed, we found that most eGFP-hPolη foci colocalized with PCNA foci (supplementary material Fig. S6). When eGFP-hPolη and TRIP-mCherry were co-expressed, a higher percentage of cells had eGFP-hPolη foci, although the average number of eGFP-hPolη foci per cell remained unchanged (Fig. 8C,D; supplementary material Fig. S7). This increased recruitment of eGFP-hPolη to nuclear foci was not secondary to increased formation of PCNA foci (Fig. 8E). Furthermore, the percentage of eGFP-hPolη foci colocalizing with PCNA foci was comparable in cells ± TRIP-mCherry co-expression, consistent with enhanced recruitment of eGFP-hPolη to existing PCNA foci rather than ectopic nuclear foci formation (supplementary material Fig. S6). Taken together, these data suggest that the TRIP/NOPO E3 ubiquitin ligase might regulate Polη by promoting its localization into PCNA foci.

We identified members of the Y-family of DNA polymerases as TRIP/NOPO interactors in yeast two-hybrid assays and co-immunoprecipitation experiments. We observed increased sensitivity to UV with nopo-like spindles and mitotic arrest of embryos from dPolη-null females. We also showed that dPolη overexpression in embryos compensates for partial loss of NOPO. We found that TRIP/NOPO enhances Polη ubiquitylation in Drosophila embryos and human cells and that TRIP promotes Polη localization to nuclear foci in human cells. Our findings support a model in which NOPO ubiquitylates dPolη during Drosophila syncytial embryogenesis to promote DNA damage tolerance in order to preserve genomic integrity, proper cell-cycle progression, and continuation of development. One limitation of the current study, however, is that most of the data supporting our model that NOPO acts upstream of dPolη were obtained from assessing the gain-of-function situation. Further studies of loss-of-function mutants of dPolη and nopo will be required to confirm our findings.

The observation that dPolη contributes to DNA damage tolerance in Drosophila embryos is consistent with previous studies in C. elegans and suggests a conserved role for Y-family DNA polymerases in ensuring cell-cycle progression during development. Germline knockdown of the dPolη homolog, polh-1, in C. elegans caused increased sensitivity of early embryos to UV irradiation, suggesting that POLH-1 is required for genome maintenance during embryogenesis (Ohkumo et al., 2006). POLH-1 has been further proposed to prevent stalling of replication forks during early embryogenesis by replacing the replicative polymerase on chromatin to quickly bypass DNA lesions (Holway et al., 2006; Kim and Michael, 2008).

Our data suggest that NOPO positively regulates dPolη activity. C. elegans POLH-1 is regulated by sumoylation, which protects it from degradation during lesion bypass (Roerink et al., 2012). POLH-1 ubiquitylation presumably occurs after it has successfully bypassed the lesion and results in ubiquitin-mediated proteolysis by the CRL4-Cdt2 E3 ligase (Kim and Michael, 2008). It will be interesting to determine whether there is a functional homolog of NOPO in C. elegans and whether Drosophila CRL4Cdt2, which regulates endoreduplication cycles by mediating E2F1 degradation, might similarly target dPolη for ubiquitin-mediated proteolysis (Zielke et al., 2011). These data would provide evidence for a conserved, highly regulated polymerase-switching model of the replicative and Y-family DNA polymerases during S phase of developing embryos.

We have shown that NOPO enhances Polη ubiquitylation during embryogenesis with reduced levels of this modification in nopo-null embryos. We did not observe decreased Polη levels by TRIP overexpression in human cells or in Drosophila wild-type versus nopo-derived embryos that would indicate TRIP/NOPO-mediated targeting of the polymerase for proteasomal degradation. This observation, along with our genetic interaction and localization data suggesting positive regulation of Polη function, argues against promotion of K48-linked poly-ubiquitylation of Polη by TRIP/NOPO E3 ligase.

We previously showed that NOPO interacts with Bendless (Ben), an E2 ubiquitin-conjugating enzyme that is the Drosophila homolog of Ubc13 (Merkle et al., 2009; Muralidhar and Thomas, 1993; Oh et al., 1994; Zhou et al., 2005). In budding yeast, the E2 heterodimer Ubc13-Mms2 mediates K63-linked poly-ubiquitylation of PCNA during postreplicative repair (Hoege et al., 2002). Mammalian Ubc13-Mms2 functions in DNA damage repair and promotion of K63 ubiquitin chain assembly (Andersen et al., 2005). Given that K63-linked ubiquitin chains generally act as nonproteolytic signals modulating protein-protein interactions and are known to facilitate DNA repair, we propose that TRIP/NOPO E3 ligase acts with Ubc13-Mms2 E2 heterodimers to mediate assembly of K63-linked poly-ubiquitin chains on Polη to facilitate bypass of DNA lesions and preserve genomic integrity in human cells and Drosophila embryos (Aguilar and Wendland, 2003; Spence et al., 1995).

Studies in budding and fission yeast and mammalian cells have established UV as a major trigger for recruiting Y-family polymerases to mono-ubiquitylated PCNA at sites of stalled replication forks (Lehmann, 2005; Watanabe et al., 2004). In unchallenged cells, mammalian Y-family polymerases localize throughout the nucleus. In response to UV, however, the polymerases are recruited to stalled replication forks and appear as foci on chromatin during S phase (Kannouche et al., 2001; Kannouche et al., 2003; Mukhopadhyay et al., 2004; Murakumo et al., 2006; Ogi et al., 2005). The mechanism by which this controlled intracellular movement of hPolη to sites of DNA damage occurs is not well understood.

The activity of hPolη and its localization to nuclear foci is inhibited by mono-ubiquitylation by the E3 ubiquitin ligase Pirh2 (Bienko et al., 2010; Jung et al., 2011). In response to UV irradiation, Protein kinase C-dependent phosphorylation of hPolη promotes hPolη recruitment to repair foci, and ATR-dependent hPolη phosphorylation contributes to efficient bypass of UV-induced DNA lesions and cell survival (Chen et al., 2008; Göhler et al., 2011). Here we show that co-expression of TRIP and hPolη in cultured human cells similarly promotes increased localization of hPolη to nuclear foci containing PCNA. We propose that TRIP/NOPO-dependent ubiquitylation of Polη results in its translocation to nuclear foci, thereby promoting its recruitment to and interaction with mono-ubiquitylated PCNA on chromatin.

The ubiquitylation state of a protein can be regulated by phosphorylation; this may occur by modulation of E3 ligase activity or facilitation of E3 ligase target recognition (Hunter, 2007). Phosphorylation can induce poly-ubiquitylation that results in proteasomal degradation of target proteins, and studies on components of the TNF-signaling pathway have shown that phosphorylation-dependent K63-linked poly-ubiquitylation can mediate protein-protein interactions or activity of target proteins (Miyamoto et al., 1998; Pickart, 1997). Determining whether TRIP/NOPO-dependent ubiquitylation of Polη and its role in promoting translocation of Polη into nuclear foci require PKC- or ATR-dependent phosphorylation will further enrich our understanding of the mechanisms by which Polη is regulated.

There is evidence that mammalian TRIP, like NOPO, is crucial during development. Mouse embryos lacking TRIP undergo early arrest with proliferation defects and excessive cell death (Park et al., 2007). TRIP also has a reported role in TNF/NF-κB-dependent sexual dimorphism in developing neurons (Krishnan et al., 2009). The authors found an upregulation of TRIP in the developing male anteroventral periventricular (AVPV) nucleus and showed that the size difference in male versus female AVPV nuclei results from apoptosis in developing male neurons (Krishnan et al., 2009). It will be interesting to determine if any of these proposed roles for TRIP in apoptosis and cell proliferation involve its regulation of Y-family DNA polymerases.

Although dPolη overexpression suppressed the partial loss of NOPO in embryos from females homozygous for a hypomorphic allele of nopo, we did not find obvious developmental defects in dPolη-derived embryos. Based on this observation, it is clear that regulation of dPolη is not the sole mechanism by which NOPO acts to preserve genomic integrity in early embryos of Drosophila. Future work will be necessary to determine whether other Y-family polymerases are involved and to identify additional targets of NOPO E3 ubiquitin ligase that are crucial for maintaining genomic stability and proper progression of development.

Drosophila stocks

y1w1118 was used as ‘wild-type’ stock. UASp-His7-Ub and mnk6006 stocks were gifts from Lynn Cooley (Yale) and Bill Theurkauf (UMass Worcester), respectively. nopoExc142 flies were previously described (Merkle et al., 2009). Other fly stocks were from Bloomington or Szeged stock centers.

cDNA clones

cDNA clones encoding NOPO, dPolη and dPolι (GH03577, SD05329 and LD29090, respectively) were from the Drosophila Gene Collection. Human TRIP cDNA (ID 2821007) was from Open Biosystems. hPolη and hPolκ clones were gifts from Peter Guengerich (Vanderbilt). Supplementary material Table S2 shows primers used for generating DNA constructs.

Transgenesis

For nopo rescue experiments, the previously described pCaSpeR4-CG5140 (containing a 3.8-kb genomic fragment including nopo and flanking regions) was used as template (Merkle et al., 2009). nopo genomic sequence between the 5′ and 3′ UTRs was replaced by nopo or human TRIP open reading frame (ORF) to generate pCaSpeR4-nopogennopocDNA or pCaSpeR4-nopogenTRIPcDNA, respectively (Fig. 1A). For dPolη rescue and overexpression experiments, cDNA was subcloned into modified UASp to generate pUASp-HA-dPolη (Rørth, 1998). Transgenic lines were generated by P-element-mediated transformation (Rubin and Spradling, 1982).

Generation of dPolη null allele

The dPolη-null allele was generated by imprecise excision of P-element EY07711. dPolηExc2.15 contains a 1309 bp deletion in the dPolη gene that removes part of the 5′ UTR and exons encoding residues 1-436.

RNA isolation and RT-PCR

RNA STAT-60 (Tel-Test) was used to extract RNA from embryos (0-2 hour). Reverse transcription was performed using the High Capacity cDNA RT Kit (Applied Biosystems) and reaction products used as PCR templates. Supplementary material Table S1 shows RT-PCR primers. PCR products were resolved by agarose gel electrophoresis. For quantitative RT-PCR, RNA was isolated from embryos, third instar larvae, pupae, ovaries and testes of adult flies (2-3 day), and carcasses (minus gonads) of adult flies for cDNA synthesis as described above. Real-time PCR was performed using RT2 SYBR Green w/Fluorescein qPCR Mastermix (Qiagen) on the Bio-Rad CFX96 Real-Time PCR system. RP49 was used as internal control. Relative transcript abundance was calculated using the ΔΔCt method. Three independent RNA samples were prepared per genotype. P-values were determined using a two-tailed, unpaired Student’s t-test.

Quantification of hatch rates

Hatch rates (eggs to larvae) were determined as previously described (Merkle et al., 2009). Hatch rate is the ratio of hatched eggs to total eggs laid expressed as a percentage. Two hundred or more embryos were scored per genotype. P-values were determined using Fisher’s exact test.

Yeast two-hybrid screen

The cDNA library, protocols and reagents were from Clontech. Human TRIP cDNA was subcloned into pGBKT7 (bait vector) to encode a fusion protein containing the Gal4 DNA-binding domain. Yeast strain AH109 was transformed with pGBKT7-hTRIP, and fusion protein expression was confirmed by immunoblotting. The bait strain was mated with yeast pre-transformed with a HeLa cDNA library. Transformants with positive interactions were selected on minimal media lacking Leu, Trp and His (triple dropout, TDO). Positive prey plasmids were sequenced and retested on TDO selection medium to confirm interactions.

Yeast two-hybrid assays

Protocols and reagents for yeast two-hybrid assays were from Clontech. cDNAs encoding full-length human TRIP or Drosophila NOPO were subcloned into pGBKT7 (bait vector) and transformed into strain AH109. cDNAs encoding full-length or truncated forms of hPolη, hPolκ, dPolη or dPolι were subcloned into pGADT7 (prey vector) and transformed into strain Y187. After mating, diploid cells co-expressing bait and prey constructs were selected by growth on media lacking Leu and Trp (double dropout, DDO). Interactions were tested by spotting cells onto TDO medium (described above) and scoring growth after 3 days at 30°C. Assays were performed three or more times with plating in triplicate.

Cell culture and transfection

HeLa cells were maintained at 37°C and 5% CO2 in Dulbecco’s modified Eagle medium (DMEM) containing 10% fetal bovine serum (FBS), 1% L-glutamine, 100 μg/ml streptomycin, and 100 U/ml penicillin (Gibco). Plasmids made by subcloning cDNAs into tagged pCS2 vectors (2 μg each) were transfected into cells using Lipofectamine 2000 (Invitrogen) for co-immunoprecipitations or Fugene HD (Promega) for immunostaining according to manufacturers’ instructions.

Drosophila S2 cells were maintained at 28°C in Schneider’s medium containing 10% FBS, 100 μg/ml streptomycin, and 100 U/ml penicillin (Gibco). Plasmids generated by subcloning cDNAs into tagged pRmHa3 vectors (2 μg each) were transfected into cells with calcium phosphate using standard methods. At 24 hours post-transfection, cells were CuSO4-treated (0.5 M) for 24 hours to induce expression.

Co-immunoprecipitation and immunoblotting

HeLa or S2 cells (24 hours post-transfection or post-induction, respectively) were lysed in nondenaturing lysis buffer (NDLB) (50 mM Tris-Cl pH 7.4, 300 mM NaCl, 5 mM EDTA, 1% Triton X-100, protease inhibitors) and centrifuged 10 minutes at 13,000 rpm. For co-immunoprecipitations, lysates (500 μg) were incubated 2 hours with shaking at 4°C with 50 μl of Ultralink Biosupport resin (Thermo Scientific) coupled to anti-MYC (9E10) or anti-FLAG (M2) antibodies. Beads were washed three times with NDLB. Bound proteins and lysates were analyzed by immunoblotting with the following primary antibodies: anti-FLAG (M2, Sigma, 1:1000), anti-c-MYC (9E10, 1:2500), and anti-HA (3F10, 1:500, Roche). Secondary antibodies were conjugated to horseradish peroxidase (HRP). Experiments were performed three or more times with representative results shown.

Drosophila embryos were dechorionated in 50% bleach, washed with water, and lysed in NDLB by homogenization with a pestle. Lysates were centrifuged 10 minutes at 13,000 rpm and analyzed by immunoblotting. Anti-NOPO antibodies were used as previously described (Merkle et al., 2009). Experiments were performed three or more times with representative results shown.

Immunostaining and microscopy

Drosophila embryos (0-2 hours unless otherwise indicated) were collected and dechorionated in 50% bleach (Rothwell and Sullivan, 2000). For UV treatment, dechorionated embryos were exposed to 500 J/m2 UV (254 nm) in a Stratalinker 1800 (Stratagene) and aged 30 minutes in darkness at 25°C before fixation. Embryos were fixed and devitellinized by shaking in methanol/heptane (1:1) and incubated overnight at 4°C in primary antibodies: anti-α-tubulin (DM1α, 1:500, Sigma), anti-Centrosomin (1:5000, gift from Bill Theurkauf, UMass Worcester), and anti-HA (12CA5, 1:2500). Secondary antibodies were conjugated to Alexa Fluor 488 or Cy5 (1:500, Invitrogen). Embryos were stained with propidium iodide and cleared (Fenger et al., 2000). Mitotic index was determined as previously described (Merkle et al., 2009). Two hundred or more embryos were scored per genotype. P-values were determined using Fisher’s exact test.

HeLa cells (24 hours post-transfection) were fixed in 4% formaldehyde [20 minutes at room temperature followed by 20 minutes in Tris-buffered saline (TBS) plus 0.05% Triton X-100] or methanol (20 minutes at -20°C followed by washing with TBS plus 0.01% Triton X-100) and blocked in 5% bovine serum albumin (BSA). The following primary antibodies were used: anti-Fibrillarin (38F3, 1:1000, Abcam; formaldehyde fixation), anti-PCNA (clone PC10, 1:1000, Biolegend; methanol fixation), and anti-γ-H2AX (clone JBW301, 1:500, Millipore, Billerica, MA; methanol fixation). Secondary antibodies were conjugated to Cy5 (1:1000, Invitrogen). Cells were mounted in ProLong Gold Antifade Reagent with 4′,6-diamidino-2-phenylindole (DAPI) (Invitrogen). For quantification of GFP-hPOLη and PCNA foci, cells with more than five foci were scored as positive. Experiments were performed three times with ≥200 cells scored per condition. P-values were determined using a two-tailed, unpaired Student’s t-test. Most images were obtained using a Nikon Eclipse 80i microscope with a CoolSNAP ES camera (Photometrics) and Plan-Apo 100×objective. Confocal images used to quantify foci were obtained with a Nikon A1RSi confocal microscope with Plan Apo 60×oil immersion objective.

DNA damage response assays

To test UV sensitivity, embryos (0-2 hour) were dechorionated, irradiated with 0, 250, 500 or 750 J/m2 UV (254 nm) in a Stratalinker 1800 (Stratagene), and kept in darkness at 25°C before assessing hatch rates. Larval sensitivity to HU was tested as previously described (Rickmyre et al., 2007).

Ubiquitylation assays

For HeLa cell studies, we used an established His6-ubiquitin method (Campanero and Flemington, 1997; Treier et al., 1994). pMT107 encoding His6-human ubiquitin was a gift from Bill Tansey (Vanderbilt) (Treier et al., 1994). Twenty-four hours post-transfection, cells were lysed in denaturing buffer (6 M guanidine-HCl, 0.1 M Na2HPO4/NaH2PO4, 10 mM imidazole) and histidine-tagged proteins purified using Ni-NTA beads (Qiagen) as described (Campanero and Flemington, 1997). Bound proteins and lysates were analyzed by immunoblotting. Experiments were performed three or more times with representative results shown.

For Drosophila embryo studies, we used wild-type or nopoExc142 females carrying transgenes UASp-HA-dPolη plus or minus UASp-His7-Ubiquitin (gift of Lynn Cooley, Yale) in combination with nanos-Gal4:VP16 to drive germline expression. Embryo (0-2 hour) lysates were prepared under denaturing conditions, histidine-tagged proteins purified, and bound proteins and lysates analyzed by immunoblotting. Experiments were performed five or more times with representative results shown.

We thank Lynn Cooley, Peter Guengerich, Bill Tansey and Bill Theurkauf for providing fly stocks, antibodies and plasmids. We thank Jeanne Jodoin for critical reading of the manuscript.

Author contributions

H.A.W., J.A.M. and L.A.L. conceived and designed experiments and analyzed data with intellectual contributions from E.L. and G.B.; experiments were performed by H.A.W. and J.A.M. with assistance from M.C.Y. and T.G.B. in the labs of L.A.L., E.L. and G.B.; H.A.W. and J.A.M. co-wrote the manuscript with guidance from L.A.L.

Funding

This work was supported by grants from the National Institutes of Health [GM074044 to L.A.L.; GM103926 and GM081635 to E.L.; GM069462 to G.B.; and 5T32 CA11992504 to H.A.W.]. Deposited in PMC for release after 12 months.

Abdu
U.
,
Brodsky
M.
,
Schüpbach
T.
(
2002
).
Activation of a meiotic checkpoint during Drosophila oogenesis regulates the translation of Gurken through Chk2/Mnk
.
Curr. Biol.
12
,
1645
1651
.
Abraham
R. T.
(
2001
).
Cell cycle checkpoint signaling through the ATM and ATR kinases
.
Genes Dev.
15
,
2177
2196
.
Acconcia
F.
,
Sigismund
S.
,
Polo
S.
(
2009
).
Ubiquitin in trafficking: the network at work
.
Exp. Cell Res.
315
,
1610
1618
.
Aguilar
R. C.
,
Wendland
B.
(
2003
).
Ubiquitin: not just for proteasomes anymore
.
Curr. Opin. Cell Biol.
15
,
184
190
.
Al-Hakim
A.
,
Escribano-Diaz
C.
,
Landry
M. C.
,
O’Donnell
L.
,
Panier
S.
,
Szilard
R. K.
,
Durocher
D.
(
2010
).
The ubiquitous role of ubiquitin in the DNA damage response
.
DNA Repair (Amst.)
9
,
1229
1240
.
Andersen
P. L.
,
Zhou
H.
,
Pastushok
L.
,
Moraes
T.
,
McKenna
S.
,
Ziola
B.
,
Ellison
M. J.
,
Dixit
V. M.
,
Xiao
W.
(
2005
).
Distinct regulation of Ubc13 functions by the two ubiquitin-conjugating enzyme variants Mms2 and Uev1A
.
J. Cell Biol.
170
,
745
755
.
Besse
A.
,
Campos
A. D.
,
Webster
W. K.
,
Darnay
B. G.
(
2007
).
TRAF-interacting protein (TRIP) is a RING-dependent ubiquitin ligase
.
Biochem. Biophys. Res. Commun.
359
,
660
664
.
Bienko
M.
,
Green
C. M.
,
Sabbioneda
S.
,
Crosetto
N.
,
Matic
I.
,
Hibbert
R. G.
,
Begovic
T.
,
Niimi
A.
,
Mann
M.
,
Lehmann
A. R.
, et al. 
. (
2010
).
Regulation of translesion synthesis DNA polymerase eta by monoubiquitination
.
Mol. Cell
37
,
396
407
.
Brodsky
M. H.
,
Weinert
B. T.
,
Tsang
G.
,
Rong
Y. S.
,
McGinnis
N. M.
,
Golic
K. G.
,
Rio
D. C.
,
Rubin
G. M.
(
2004
).
Drosophila melanogaster MNK/Chk2 and p53 regulate multiple DNA repair and apoptotic pathways following DNA damage
.
Mol. Cell. Biol.
24
,
1219
1231
.
Broemer
M.
,
Meier
P.
(
2009
).
Ubiquitin-mediated regulation of apoptosis
.
Trends Cell Biol.
19
,
130
140
.
Budirahardja
Y.
,
Gönczy
P.
(
2009
).
Coupling the cell cycle to development
.
Development
136
,
2861
2872
.
Campanero
M. R.
,
Flemington
E. K.
(
1997
).
Regulation of E2F through ubiquitin-proteasome-dependent degradation: stabilization by the pRB tumor suppressor protein
.
Proc. Natl. Acad. Sci. USA
94
,
2221
2226
.
Chen
Y. W.
,
Cleaver
J. E.
,
Hatahet
Z.
,
Honkanen
R. E.
,
Chang
J. Y.
,
Yen
Y.
,
Chou
K. M.
(
2008
).
Human DNA polymerase eta activity and translocation is regulated by phosphorylation
.
Proc. Natl. Acad. Sci. USA
105
,
16578
16583
.
Deshaies
R. J.
,
Joazeiro
C. A.
(
2009
).
RING domain E3 ubiquitin ligases
.
Annu. Rev. Biochem.
78
,
399
434
.
Fang
S.
,
Weissman
A. M.
(
2004
).
A field guide to ubiquitylation
.
Cell. Mol. Life Sci.
61
,
1546
1561
.
Fenger
D. D.
,
Carminati
J. L.
,
Burney-Sigman
D. L.
,
Kashevsky
H.
,
Dines
J. L.
,
Elfring
L. K.
,
Orr-Weaver
T. L.
(
2000
).
PAN GU: a protein kinase that inhibits S phase and promotes mitosis in early Drosophila development
.
Development
127
,
4763
4774
.
Fogarty
P.
,
Kalpin
R. F.
,
Sullivan
W.
(
1994
).
The Drosophila maternal-effect mutation grapes causes a metaphase arrest at nuclear cycle 13
.
Development
120
,
2131
2142
.
Freudenthal
B. D.
,
Gakhar
L.
,
Ramaswamy
S.
,
Washington
M. T.
(
2010
).
Structure of monoubiquitinated PCNA and implications for translesion synthesis and DNA polymerase exchange
.
Nat. Struct. Mol. Biol.
17
,
479
484
.
Glickman
M. H.
,
Ciechanover
A.
(
2002
).
The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construction
.
Physiol. Rev.
82
,
373
428
.
Göhler
T.
,
Sabbioneda
S.
,
Green
C. M.
,
Lehmann
A. R.
(
2011
).
ATR-mediated phosphorylation of DNA polymerase η is needed for efficient recovery from UV damage
.
J. Cell Biol.
192
,
219
227
.
Ha
H.
,
Han
D.
,
Choi
Y.
(
2009
).
TRAF-mediated TNFR-family signaling
.
Curr. Protoc. Immunol.
Chapter 11
,
Unit11 9D
.
Hershko
A.
(
1997
).
Roles of ubiquitin-mediated proteolysis in cell cycle control
.
Curr. Opin. Cell Biol.
9
,
788
799
.
Hoege
C.
,
Pfander
B.
,
Moldovan
G. L.
,
Pyrowolakis
G.
,
Jentsch
S.
(
2002
).
RAD6-dependent DNA repair is linked to modification of PCNA by ubiquitin and SUMO
.
Nature
419
,
135
141
.
Holway
A. H.
,
Kim
S. H.
,
La Volpe
A.
,
Michael
W. M.
(
2006
).
Checkpoint silencing during the DNA damage response in Caenorhabditis elegans embryos
.
J. Cell Biol.
172
,
999
1008
.
Hunter
T.
(
2007
).
The age of crosstalk: phosphorylation, ubiquitination, and beyond
.
Mol. Cell
28
,
730
738
.
Ishikawa
T.
,
Uematsu
N.
,
Mizukoshi
T.
,
Iwai
S.
,
Iwasaki
H.
,
Masutani
C.
,
Hanaoka
F.
,
Ueda
R.
,
Ohmori
H.
,
Todo
T.
(
2001
).
Mutagenic and nonmutagenic bypass of DNA lesions by Drosophila DNA polymerases dpoleta and dpoliota
.
J. Biol. Chem.
276
,
15155
15163
.
Johnson
R. E.
,
Kondratick
C. M.
,
Prakash
S.
,
Prakash
L.
(
1999
).
hRAD30 mutations in the variant form of xeroderma pigmentosum
.
Science
285
,
263
265
.
Jung
Y. S.
,
Liu
G.
,
Chen
X.
(
2010
).
Pirh2 E3 ubiquitin ligase targets DNA polymerase eta for 20S proteasomal degradation
.
Mol. Cell. Biol.
30
,
1041
1048
.
Jung
Y. S.
,
Hakem
A.
,
Hakem
R.
,
Chen
X.
(
2011
).
Pirh2 E3 ubiquitin ligase monoubiquitinates DNA polymerase eta to suppress translesion DNA synthesis
.
Mol. Cell. Biol.
31
,
3997
4006
.
Kane
D. P.
,
Shusterman
M.
,
Rong
Y.
,
McVey
M.
(
2012
).
Competition between replicative and translesion polymerases during homologous recombination repair in Drosophila
.
PLoS Genet.
8
,
e1002659
.
Kannouche
P.
,
Lehmann
A.
(
2006
).
Localization of Y-family polymerases and the DNA polymerase switch in mammalian cells
.
Methods Enzymol.
408
,
407
415
.
Kannouche
P.
,
Stary
A.
(
2003
).
Xeroderma pigmentosum variant and error-prone DNA polymerases
.
Biochimie
85
,
1123
1132
.
Kannouche
P.
,
Broughton
B. C.
,
Volker
M.
,
Hanaoka
F.
,
Mullenders
L. H.
,
Lehmann
A. R.
(
2001
).
Domain structure, localization, and function of DNA polymerase eta, defective in xeroderma pigmentosum variant cells
.
Genes Dev.
15
,
158
172
.
Kannouche
P.
,
Fernández de Henestrosa
A. R.
,
Coull
B.
,
Vidal
A. E.
,
Gray
C.
,
Zicha
D.
,
Woodgate
R.
,
Lehmann
A. R.
(
2002
).
Localization of DNA polymerases eta and iota to the replication machinery is tightly co-ordinated in human cells
.
EMBO J.
21
,
6246
6256
.
Kannouche
P.
,
Fernández de Henestrosa
A. R.
,
Coull
B.
,
Vidal
A. E.
,
Gray
C.
,
Zicha
D.
,
Woodgate
R.
,
Lehmann
A. R.
(
2003
).
Localization of DNA polymerases eta and iota to the replication machinery is tightly co-ordinated in human cells
.
EMBO J.
22
,
1223
1233
.
Kannouche
P. L.
,
Wing
J.
,
Lehmann
A. R.
(
2004
).
Interaction of human DNA polymerase eta with monoubiquitinated PCNA: a possible mechanism for the polymerase switch in response to DNA damage
.
Mol. Cell
14
,
491
500
.
Kim
S. H.
,
Michael
W. M.
(
2008
).
Regulated proteolysis of DNA polymerase eta during the DNA-damage response in C. elegans
.
Mol. Cell
32
,
757
766
.
Krishnan
S.
,
Intlekofer
K. A.
,
Aggison
L. K.
,
Petersen
S. L.
(
2009
).
Central role of TRAF-interacting protein in a new model of brain sexual differentiation
.
Proc. Natl. Acad. Sci. USA
106
,
16692
16697
.
Le Bras
S.
,
Loyer
N.
,
Le Borgne
R.
(
2011
).
The multiple facets of ubiquitination in the regulation of notch signaling pathway
.
Traffic
12
,
149
161
.
Lee
S. Y.
,
Lee
S. Y.
,
Choi
Y.
(
1997
).
TRAF-interacting protein (TRIP): a novel component of the tumor necrosis factor receptor (TNFR)- and CD30-TRAF signaling complexes that inhibits TRAF2-mediated NF-kappaB activation
.
J. Exp. Med.
185
,
1275
1286
.
Lehmann
A. R.
(
2005
).
Replication of damaged DNA by translesion synthesis in human cells
.
FEBS Lett.
579
,
873
876
.
Lehmann
A. R.
,
Niimi
A.
,
Ogi
T.
,
Brown
S.
,
Sabbioneda
S.
,
Wing
J. F.
,
Kannouche
P. L.
,
Green
C. M.
(
2007
).
Translesion synthesis: Y-family polymerases and the polymerase switch
.
DNA Repair (Amst.)
6
,
891
899
.
Masrouha
N.
,
Yang
L.
,
Hijal
S.
,
Larochelle
S.
,
Suter
B.
(
2003
).
The Drosophila chk2 gene loki is essential for embryonic DNA double-strand-break checkpoints induced in S phase or G2
.
Genetics
163
,
973
982
.
Masutani
C.
,
Kusumoto
R.
,
Yamada
A.
,
Dohmae
N.
,
Yokoi
M.
,
Yuasa
M.
,
Araki
M.
,
Iwai
S.
,
Takio
K.
,
Hanaoka
F.
(
1999
).
The XPV (xeroderma pigmentosum variant) gene encodes human DNA polymerase eta
.
Nature
399
,
700
704
.
McCulloch
S. D.
,
Kokoska
R. J.
,
Masutani
C.
,
Iwai
S.
,
Hanaoka
F.
,
Kunkel
T. A.
(
2004
).
Preferential cis-syn thymine dimer bypass by DNA polymerase eta occurs with biased fidelity
.
Nature
428
,
97
100
.
McDonald
J. P.
,
Levine
A. S.
,
Woodgate
R.
(
1997
).
The Saccharomyces cerevisiae RAD30 gene, a homologue of Escherichia coli dinB and umuC, is DNA damage inducible and functions in a novel error-free postreplication repair mechanism
.
Genetics
147
,
1557
1568
.
Merkle
J. A.
,
Rickmyre
J. L.
,
Garg
A.
,
Loggins
E. B.
,
Jodoin
J. N.
,
Lee
E.
,
Wu
L. P.
,
Lee
L. A.
(
2009
).
no poles encodes a predicted E3 ubiquitin ligase required for early embryonic development of Drosophila
.
Development
136
,
449
459
.
Miyamoto
S.
,
Seufzer
B. J.
,
Shumway
S. D.
(
1998
).
Novel IkappaB alpha proteolytic pathway in WEHI231 immature B cells
.
Mol. Cell. Biol.
18
,
19
29
.
Mukhopadhyay
S.
,
Clark
D. R.
,
Watson
N. B.
,
Zacharias
W.
,
McGregor
W. G.
(
2004
).
REV1 accumulates in DNA damage-induced nuclear foci in human cells and is implicated in mutagenesis by benzo[a]pyrenediolepoxide
.
Nucleic Acids Res.
32
,
5820
5826
.
Murakumo
Y.
,
Mizutani
S.
,
Yamaguchi
M.
,
Ichihara
M.
,
Takahashi
M.
(
2006
).
Analyses of ultraviolet-induced focus formation of hREV1 protein
.
Genes Cells
11
,
193
205
.
Muralidhar
M. G.
,
Thomas
J. B.
(
1993
).
The Drosophila bendless gene encodes a neural protein related to ubiquitin-conjugating enzymes
.
Neuron
11
,
253
266
.
O’Connell
B. C.
,
Harper
J. W.
(
2007
).
Ubiquitin proteasome system (UPS): what can chromatin do for you?
Curr. Opin. Cell Biol.
19
,
206
214
.
O’Farrell
P. H.
,
Stumpff
J.
,
Su
T. T.
(
2004
).
Embryonic cleavage cycles: how is a mouse like a fly?
Curr. Biol.
14
,
R35
R45
.
Ogi
T.
,
Kannouche
P.
,
Lehmann
A. R.
(
2005
).
Localisation of human Y-family DNA polymerase kappa: relationship to PCNA foci
.
J. Cell Sci.
118
,
129
136
.
Oh
C. E.
,
McMahon
R.
,
Benzer
S.
,
Tanouye
M. A.
(
1994
).
bendless, a Drosophila gene affecting neuronal connectivity, encodes a ubiquitin-conjugating enzyme homolog
.
J. Neurosci.
14
,
3166
3179
.
Ohkumo
T.
,
Masutani
C.
,
Eki
T.
,
Hanaoka
F.
(
2006
).
Deficiency of the Caenorhabditis elegans DNA polymerase eta homologue increases sensitivity to UV radiation during germ-line development
.
Cell Struct. Funct.
31
,
29
37
.
Park
E. S.
,
Choi
S.
,
Kim
J. M.
,
Jeong
Y.
,
Choe
J.
,
Park
C. S.
,
Choi
Y.
,
Rho
J.
(
2007
).
Early embryonic lethality caused by targeted disruption of the TRAF-interacting protein (TRIP) gene
.
Biochem. Biophys. Res. Commun.
363
,
971
977
.
Pickart
C. M.
(
1997
).
Targeting of substrates to the 26S proteasome
.
FASEB J.
11
,
1055
1066
.
Regamey
A.
,
Hohl
D.
,
Liu
J. W.
,
Roger
T.
,
Kogerman
P.
,
Toftgard
R.
,
Huber
M.
(
2003
).
The tumor suppressor CYLD interacts with TRIP and regulates negatively nuclear factor kappaB activation by tumor necrosis factor
.
J. Exp. Med.
198
,
1959
1964
.
Rey
L.
,
Sidorova
J. M.
,
Puget
N.
,
Boudsocq
F.
,
Biard
D. S.
,
Monnat
R. J.
Jr
,
Cazaux
C.
,
Hoffmann
J. S.
(
2009
).
Human DNA polymerase eta is required for common fragile site stability during unperturbed DNA replication
.
Mol. Cell. Biol.
29
,
3344
3354
.
Rickmyre
J. L.
,
Dasgupta
S.
,
Ooi
D. L.
,
Keel
J.
,
Lee
E.
,
Kirschner
M. W.
,
Waddell
S.
,
Lee
L. A.
(
2007
).
The Drosophila homolog of MCPH1, a human microcephaly gene, is required for genomic stability in the early embryo
.
J. Cell Sci.
120
,
3565
3577
.
Roerink
S. F.
,
Koole
W.
,
Stapel
L. C.
,
Romeijn
R. J.
,
Tijsterman
M.
(
2012
).
A broad requirement for TLS polymerases η and κ, and interacting sumoylation and nuclear pore proteins, in lesion bypass during C. elegans embryogenesis
.
PLoS Genet.
8
,
e1002800
.
Rørth
P.
(
1998
).
Gal4 in the Drosophila female germline
.
Mech. Dev.
78
,
113
118
.
Rothwell
W. F.
,
Sullivan
W.
(
2000
).
Fluorescent analysis of Drosophila embryos
. In
Drosophila Protocols
(ed.
Sullivan
W.
,
Ashburner
M.
,
Hawley
R. S.
), pp.
141
157
.
Cold Spring Harbor, NY
:
Cold Spring Harbor Laboratory Press
.
Rubin
G. M.
,
Spradling
A. C.
(
1982
).
Genetic transformation of Drosophila with transposable element vectors
.
Science
218
,
348
353
.
Sale
J. E.
,
Lehmann
A. R.
,
Woodgate
R.
(
2012
).
Y-family DNA polymerases and their role in tolerance of cellular DNA damage
.
Nat. Rev. Mol. Cell Biol.
13
,
141
152
.
Sancar
A.
,
Lindsey-Boltz
L. A.
,
Unsal-Kaçmaz
K.
,
Linn
S.
(
2004
).
Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints
.
Annu. Rev. Biochem.
73
,
39
85
.
Shaheen
M.
,
Shanmugam
I.
,
Hromas
R.
(
2010
).
The role of PCNA posttranslational modifications in translesion synthesis
.
J. Nucleic Acids
2010
,
761217
.
Sibon
O. C.
,
Stevenson
V. A.
,
Theurkauf
W. E.
(
1997
).
DNA-replication checkpoint control at the Drosophila midblastula transition
.
Nature
388
,
93
97
.
Sibon
O. C.
,
Laurençon
A.
,
Hawley
R.
,
Theurkauf
W. E.
(
1999
).
The Drosophila ATM homologue Mei-41 has an essential checkpoint function at the midblastula transition
.
Curr. Biol.
9
,
302
312
.
Sibon
O. C.
,
Kelkar
A.
,
Lemstra
W.
,
Theurkauf
W. E.
(
2000
).
DNA-replication/DNA-damage-dependent centrosome inactivation in Drosophila embryos
.
Nat. Cell Biol.
2
,
90
95
.
Skoneczna
A.
,
McIntyre
J.
,
Skoneczny
M.
,
Policinska
Z.
,
Sledziewska-Gojska
E.
(
2007
).
Polymerase eta is a short-lived, proteasomally degraded protein that is temporarily stabilized following UV irradiation in Saccharomyces cerevisiae
.
J. Mol. Biol.
366
,
1074
1086
.
Spence
J.
,
Sadis
S.
,
Haas
A. L.
,
Finley
D.
(
1995
).
A ubiquitin mutant with specific defects in DNA repair and multiubiquitination
.
Mol. Cell. Biol.
15
,
1265
1273
.
Takada
S.
,
Kelkar
A.
,
Theurkauf
W. E.
(
2003
).
Drosophila checkpoint kinase 2 couples centrosome function and spindle assembly to genomic integrity
.
Cell
113
,
87
99
.
Treier
M.
,
Staszewski
L. M.
,
Bohmann
D.
(
1994
).
Ubiquitin-dependent c-Jun degradation in vivo is mediated by the delta domain
.
Cell
78
,
787
798
.
Watanabe
K.
,
Tateishi
S.
,
Kawasuji
M.
,
Tsurimoto
T.
,
Inoue
H.
,
Yamaizumi
M.
(
2004
).
Rad18 guides poleta to replication stalling sites through physical interaction and PCNA monoubiquitination
.
EMBO J.
23
,
3886
3896
.
Waters
L. S.
,
Minesinger
B. K.
,
Wiltrout
M. E.
,
D’Souza
S.
,
Woodruff
R. V.
,
Walker
G. C.
(
2009
).
Eukaryotic translesion polymerases and their roles and regulation in DNA damage tolerance
.
Microbiol. Mol. Biol. Rev.
73
,
134
154
.
Xu
J.
,
Xin
S.
,
Du
W.
(
2001
).
Drosophila Chk2 is required for DNA damage-mediated cell cycle arrest and apoptosis
.
FEBS Lett.
508
,
394
398
.
Zhou
Q.
,
Geahlen
R. L.
(
2009
).
The protein-tyrosine kinase Syk interacts with TRAF-interacting protein TRIP in breast epithelial cells
.
Oncogene
28
,
1348
1356
.
Zhou
R.
,
Silverman
N.
,
Hong
M.
,
Liao
D. S.
,
Chung
Y.
,
Chen
Z. J.
,
Maniatis
T.
(
2005
).
The role of ubiquitination in Drosophila innate immunity
.
J. Biol. Chem.
280
,
34048
34055
.
Zielke
N.
,
Kim
K. J.
,
Tran
V.
,
Shibutani
S. T.
,
Bravo
M. J.
,
Nagarajan
S.
,
van Straaten
M.
,
Woods
B.
,
von Dassow
G.
,
Rottig
C.
, et al. 
. (
2011
).
Control of Drosophila endocycles by E2F and CRL4(CDT2)
.
Nature
480
,
123
127
.

Competing interests

The authors declare no competing financial interests.

Supplementary information