DNA LIGASE IV STRUCTURALLY SUPPORTS END JOINING REPAIR OF DNA DOUBLE STRAND BREAKS. By Noah J. Goff A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of Cell and Molecular Biology – Doctor of Philosophy 2024 ABSTRACT DNA Double strand breaks (DSBs) are highly genotoxic lesions induced by external agents (ionizing radiation or chemotherapeutic drugs), cellular processes (DNA replication defects, reactive oxygen species, recombination intermediates in B/T cell development), or through increasingly prevalent gene editing techniques (CRISPR/Cas9). Mammals have evolved two major pathways for repairing DSBs: Homologous Recombination (HR) and Non-Homologous End Joining (NHEJ). While NHEJ is frequently referred to as the “error prone” pathway, recent biochemical and in vitro single-molecule imaging studies have revealed mechanisms to protect ends from excessive end-processing through a two-stage synaptic model. First, DNA ends are synapsed by NHEJ factors in a long-range complex (LRC) with ends held ~115 Å apart. The LRC recruits downstream factors and only permits highly regulated end processing of bulky adducts. Second— following the recruitment of the NHEJ-associated DNA Ligase IV (L4)—ends are brought into direct proximity for final end-processing and ligation. In this dissertation, I report that catalytically inactive L4 promotes significant amounts of end joining in cell models, supporting the recent two- stage synaptic model for NHEJ. Furthermore, I characterize repair products from cells expressing catalytically inactive L4 showing that repair is significantly more mutagenic than in cells expressing active L4. Finally, I identify multiple interfaces in L4’s DNA binding domain critical for maintaining promoting repair, providing insight into how L4 structurally supports synapsis of ends. This dissertagon is dedicated to my parents. It’s impossible to overstate the significance of your support over the past 5 years. iii ACKNOWLEDGMENTS The past five years of research and gme spent wrigng for my PhD would not have been possible without the generous and thoughiul support from dozens of people. It absolutely takes a village and I’m extremely grateful to the amazing community of people who have helped me over the past ~5 years. Special thanks to my commijee for your valuable advice and well-thought quesgons throughout my gme here at MSU; Dr. Daniel Vocelle from the MSU Flow Cytometry core for your dedicagon to teaching and technical know-how; the friends I’ve made through the BMS and CMB programs for their willingness to discuss science at any gme of day (preferably over a beer); Dr. Jens Schmidt’s lab for being so accommodagng with microscope gme (and room in their biosafety cabinets); the CMB program—specifically Dr. Margaret Petroff for being a fantasgc graduate director and Alaina Burghardt for being the best program coordinator at the university; Emily Durocher for her unerring ability to make me smile amer a long day; and finally my family for being there since day one, I’m so happy with how this journey has turned out and It’s impossible to quangfy how much having you nearby has meant along the way (not to mengon all of the free food). iv TABLE OF CONTENTS Chapter 1: Evolving models of non-homologous end joining. ........................................................ 1 REFERENCES ..................................................................................................................... 18 Chapter 2: Catalygcally inacgve DNA Ligase IV promotes DNA repair in living cells. ................. .25 REFERENCES ..................................................................................................................... 51 Chapter 3: New insight into how the DNA binding domain of DNA Ligase IV facilitates end- joining, independent of its catalygc acgvity. ............................................................................... 56 REFERENCES ..................................................................................................................... 78 Chapter 4: Discussing the two-stage synapgc model of NHEJ. .................................................... 81 REFERENCES ..................................................................................................................... 95 APPENDIX: A list of authorship contribugons to other papers. ....................................... 97 v Chapter 1: Evolving models of non-homologous end joining. By Noah J. Goff Introducgon and literature review 1 INTRODUCTION DNA double strand breaks (DSBs) are extremely genotoxic lesions generated by a host of cellular processes and exogenous agents. Repair of DSBs is essengal, and consequences of unrepaired breaks can impair core nuclear funcgons (transcripgon, DNA replicagon), stall cell cycle progression by triggering DNA damage checkpoints (DSB forming drugs form the backbone of many chemotherapeugc treatments), compromise genomic stability through aberrant genomic translocagons, and ulgmately an unrepaired break can lead to programmed cell death. Furthermore, failures during repair processes can further compromise genomic integrity, including short insergons/delegons, copy number variagons, large intrachromosomal delegons, and inappropriate pairing of ends can exacerbate large chromosomal delegons. My work has focused on DNA end joining pathways and the structural role that DNA Ligase IV (L4) plays in supporgng end joining through the canonical non-homologous end joining pathway (NHEJ) and how loss of L4 catalygc acgvity promotes mutagenic alternagve end joining (alt-EJ). DSB Repair pathways aim to accurately resolve breaks. DSBs are generally repaired through two mechanisms: NHEJ and homologous recombinagon (HR). Briefly (DSB repair pathways are discussed in greater detail later in this chapter), HR uglizes a nearby homologous DNA sequence to perform high fidelity templated repair. NHEJ serves to resolve DSB repair through direct ligagon of two free DNA ends (hence “end joining”). First, a DNA end joining pathway must sense free DNA ends, then generate a ligagon substrate by modifying chemistry at the ends, and eventually catalygcally seal the break by ligagng both broken phosphodiester backbones. NHEJ solves a core challenge for cells: how to repair breaks that occur without a nearby homologous template (which is only present immediately following DNA replicagon). The flexibility of NHEJ (it can act on DNA ends through nearly the engre cell cycle) presents several challenges; namely, how to conserve sequence idengty (prevengng indels) and to conserve larger chromosomal structures (prevengng large genomic translocagons). Recent work has highlighted the remarkable fidelity of NHEJ and has sought to idengfy mechanisms to minimize errors generated by repair. Conserved NHEJ structural factors promote repair. A structural heterodimer formed by Ku70 and Ku80 serves both as the key inigal sensor of DNA damage as well as an anchor point for downstream factor recruitment. Early studies into Ku revealed that each factor exists 2 independently of each other but rapidly bind to DSBs and promote recruitment of other factors (citagons). The two “C” shaped proteins clamp onto a double strand break, forming a donut shape that mediates protein-protein interacgons with DNA-PKcs (forming the DNA-PK holoenzyme) and other repair factors (frequently though interacgons between Ku binding mogfs (KBM) and BRCT domains on other repair factors). In human cell strains, loss of either Ku gene is lethal, resulgng in telomere fusions and cell death (1, 2). Interesgngly, the heterodimeric ring formed by Ku70/Ku80 is extremely stable and allowed to slide upstream of the break site—like beads on a string. As such, several mechanisms exist to prevent overloading of Ku onto a DNA an end and there’s significant discussion and ongoing research into how these gghtly-bound DNA repair donuts are removed from repaired ends (3, 4). Several structural features of Ku have been well characterized, including the Ku80 C- Terminal Domain (in pargcular the final 14 residues) as being crigcal for funcgonal interacgons with DNA-PKcs to promote end joining (5–7). X ray cross complimengng protein 4 (XRCC4) serves both as a key structural factor in supporgng NHEJ synapsis and as the obligate binding partner of Ligase IV (L4). Two molecules of XRCC4 and one molecule of L4 form the ligagon complex LX4, together a core NHEJ factor that is necessary for robust end joining. Each XRCC4 homodimer is shaped with an N-terminal globular head and long C-terminal alpha-helix stalk that mediates its interacgon with L4 (8, 9). When homodimerized, the two XRCC4 head domains pair up with the C-terminal alpha-helices wound around each other 1-2 gmes (Figures 1, 2, 4). Loss of XRCC4 or L4 results in extreme hypersensigvity to DSB inducing agents (both as a result of radiagon and drug induced damage) (10–16). Addigonally, XRCC4 possesses a disordered c-terminal “tail” region that has been shown to transiently interact with L4’s DBD and directly with DNA (17). As part of the LX4 complex, XRCC4 facilitates interacgons with XLF and Ku, providing support for end synapsis observed in recent Cryo-EM structures (18, 19). 3 Figure 1: LX4 Has been observed in the Ku80-Mediated Long-Range Complex when incubated with PAXX. PDB 8BHY: dimeric structure of DNA-PKcs (grey), Ku70/Ku80 (grey/black), L4 (green), XRCC4 (purple), XLF (orange), and PAXX (red) work together to synapse DNA (orange) in a long- range synapse. Here, the LX4 complex are posigoned away from the break, with interacgons between DNA-PK and PAXX, along with trans interacgons between Ku80 and DNA-PKcs supporgng synapsis. Addigonal structural factors XRCC4-like factor (XLF) and paralog of XRCC4 and XLF (PAXX) also work to synapse ends. XLF is a central mediator of synapsis in recent Cryo-EM structures, with the “XLF-mediated dimer” consisgng of a group of structures where an XLF homodimer serves as a major contact in bridging the end-bound DNA-PK holoenzyme and LX4 complex (18, 19). The XLF-mediated dimer appears to fill the role of “end protecgon complex” originally proposed as the primary role of the long-range complex (LRC) proposed by Loparo and colleagues (20, 21). While complete ablagon of XLF has variable impacts in different organisms, loss of XLF is remarkably synthegcally lethal with a host of other DNA factors, and many studies implicate its role in promogng high-fidelity repair (12, 22–28). While typically not considered a “core” NHEJ factor (PAXX is funcgonally redundant with XLF), PAXX has recently been observed supporgng LRC 4 via Cryo-EM, including a structure very similar to the Ku80-mediated dimer that includes PAXX and LX4 (providing evidence that the NHEJ dimers may all be capable of recruigng core factors required for transigon into the SRC). Despite its apparent redundancy with XLF, PAXX has been idengfied in a wide range of species including plants (29). The two core enzymagc NHEJ factors, DNA-PKcs and L4, each play unique structural roles in supporgng NHEJ. DNA-PKcs is a large 465 kDa serine/threonine protein kinase that acts as the primary structural regulator of DNA end access. Furthermore, DNA-PKcs autophosphorylagon plays a key role in driving conformagon changes in NHEJ complexes, with well-studied phosphorylagon mogfs directly regulagng access to DNA ends (11, 30–32). While not present in prokaryotes (many prokaryotes have end-joining with distant orthologs of the Ku and L4 molecules) (33, 34), DNA-PKcs is found in a wide range of eukaryotes with many of its key structural mogfs (and autophosphorylagon sites) conserved (35). Mechanism and domains of mammalian DNA ligases: There are three mammalian DNA ligase genes: Ligase I (L1), Ligase III (L3) and Ligase IV (L4). All three genes share a conserved catalygc core consisgng of a reacgve Nucleogdyl Transferase Domain (NTD) flanked by a DNA-Binding Domain (DBD) and Oligonucleogde-Binding-Fold (OBF). While all three ligases serve DNA repair funcgons in the nucleus, L3 also provides mitochondrial DNA repair funcgon through an alternate translagonal start site. This replaces the extreme N-terminus of the protein, subsgtugng the nuclear localizagon signal for a mitochondrial localizagon signal (36). Each mammalian ligase is ATP dependent, capable of sealing a single nick in a deoxyribose-phosphate backbone (37–39). The general reacgon progresses in three steps: the ligase 1) hydrolyzes ATP to adenylate a conserved lysine residue in NTD acgve-site, 2) binds to a nick and transfers the AMP moiety from its lysine to the 5’ phosphate, then 3) supports a nucleophilic ajack from the remaining 3’ hydroxyl end to the 5’ PO4-AMP bond resulgng in a free AMP, uncharged ligase, and sealed single- strand break (37–41). Notably, the high degree of domain-conservagon and architecture across all three DNA ligases implies that these genes share a common evolugonary ancestor (37, 39– 41). Each Ligase NTD acts primarily through a conserved catalygc lysine that self-adenylated in the presence of ATP, providing energy to catalyze the break (37, 39). 5 Ligase IV is the NHEJ-exclusive ligase and well adapted for DSB repair. While Ligases I and III are frequently referred to as redundant, L4 serves exclusively in NHEJ-mediated DSB repair. While each of the mammalian ligases share a conserved mechanism and degree of structural homology in their catalygc cores, L4 has evolved several unique structural features to promote ligagng ends from a DSB: First, L4 has evolved a pair of tandem BRCT (BRCA1 c-terminus) domains that facilitate its interacgons with NHEJ proteins. The tandem L4 BRCT domains are connected by a short, flexible linker termed the XRCC4 Interacgng Region (XIR) for its role intercalagng into the central alpha- helix stalk of an XRCC4-homodimer (37–41). The XIR is both sufficient and necessary to mediate L4’s interacgon with XRCC4 (of note, L3 also has evolved n-terminal BRCT domains that mediate its interacgon with XRCC1). Second, unlike L1/L3, L4 rapidly adenylates following translagon, and nearly all the L4 molecules present within the nucleus are prepared to catalyze break repair (37). This readiness could serve as a mechanism to minimize the amount of gme required to perform the inigal ligagon step—convergng the DSB into a much less harmful single-stranded break. Interesgngly, while we were studying a catalygcally inacgve L4 (designed by mutagng the conserved catalygc lysine), we observed residual ligagon acgvity in in vitro joining assays. This residual ligase acgvity is lost following mutagon of addigonal lysines posigoned close to the DNA end, indicagng that there is some flexibility within the catalygc core (10). To my knowledge, there have been no reports of alternate lysine adenylagon in L1 or L3. Third, L4 has a unique 8-amino acid loop structure in its DBD that promotes flexibility in DNA-substrate binding, including tolerance of mismatches, aberrant bases, and gaps in one end of the DNA substrate (42). This unique lesion tolerance is another representagon of the priority given to double strand break repair—with failure to catalyze repair resulgng in significant cell- growth defects (and loss of L4 is generally lethal) (14, 43). Notably in cell line models, loss of L1 or nuclear L3 generally does not result in cell fitness defect or even notable drug-sensigvity phenotypes (15, 44). The two single-stranded ligases are funcgonally redundant and loss of both is synthegcally lethal; in contrast, L4 does not appear to complement DNA repair outside of NHEJ. DNA-PKcs: A Synopsis Beyond Synapsis. The following secgons have been reproduced from a review that I co-wrote with another graduate student, Maria Mikhova, alongside our advisors 6 Katheryn Meek and Jens Schmidt (45). The reproduced secgons were primarily dramed by me and edited as a group. The figure referenced in this secgon was originally produced by Dr. Meek, originally produced as as “Figure 2” within the review. I’ve also removed chapter numbers, updated citagon formats, citagon numbers, and abbreviagons to be consistent with the styles used in my dissertagon. A two-step model of DNA end synapsis in long and short-range NHEJ complexes. An emerging consensus in the field is a two-stage synapgc model for NHEJ driven by the presence and catalygc acgvity of DNA-PK. This model was inigally proposed by Loparo and colleagues who have extensively employed X. Laevis egg extracts to measure NHEJ-mediated synapsis of two DNA ends using single-molecule fluorescence resonance energy transfer (smFRET, simultaneously measuring synapsis and proximity). Graham et al. provided the first evidence and model for disgnct long-range and short-range synapgc complexes [LRC, SRC] (46). They proposed a LRC which notably holds DNA ends far apart (>100 Å); these LRCs are short-lived (on the order of several seconds), and only a subset progress to SRCs where ends are brought in direct proximity (as assessed by FRET). Formagon of LRCs requires only Ku and DNA-PKcs. Crigcally, in this study, acceptor fluorescence was visualized independently of donor-mediated FRET allowing observagon of synapgc events at distances beyond the range of FRET detecgon (>100 Å). This study established that progression to the SRC required the presence of Ku, L4, XRCC4, XLF, and the catalygc acgvity of DNA-PKcs. Interesgngly, the catalygc acgvity of LX4, essengal for the final ligagon step in NHEJ, was engrely dispensable for SRC formagon, supporgng an important structural role for LX4 which had been proposed in previous studies (12, 47). Our recent cellular studies confirmed that LX4’s catalygc acgvity is dispensable for progression from long-range to short-range complexes (10). Moreover, these findings imply that nuclear DNA ligase III (L3) can access DNA ends maintained in NHEJ complexes by the presence of catalygcally inacgve LX4, a heretofore unappreciated level of cooperagvity between mammalian DNA ligases both in living cells (10) and in animal models (14). LX4 binding to DNA ends drives progression from long-range to short-range complexes. Work from Loparo and colleagues has recently addressed how NHEJ priorigzes ligagon (of compagble ends) over end processing, promogng error-free repair. A 3-color imaging system was uglized to 7 independently assess LX4 binding at each end, the presence or absence of short-range synapsis, and LX4 stoichiometry, independent of its end-binding (48). This study demonstrates that LX4 dynamically binds DNA ends through residues in its DNA Binding Domain (DBD) prior to short- range synapsis, potengally as a sensor for large end adducts that could impede ligagon or final end-processing. SRC assembly (as measured by FRET between DNA ends) occurs exactly at the gme when a single LX4 complex binds to the two DNA ends. Surprisingly, SRC formagon results in evicgon of one of the two LX4 complexes that are present prior to short-range synapsis. Immediately at the gme of short-range synapsis, loss of one LX4 complex was observed with the second LX4 residing for much longer consistent with the previously reported persistence of short- range complexes. A caveat of this study is that a blunt-ended DNA substrate was uglized that should be readily ligated, although LX4 end-binding progression to a short-range complex was similarly efficient with or without 5’ phosphates. An important quesgon remains as to whether the change in LX4 stoichiometry occurs prior to strand ligagon. A similarly important issue is determining how LX4’s dwell gme is impacted if the DNA ends require fill-in synthesis or nucleolygc processing on one or both ends. An ajracgve hypothesis supported by robust previous studies (49, 50) would be that the immediate capacity to ligate paired ends of one of the DNA strands would funcgon even if the second strand requires substangal end-processing. Simply put, there is no bejer way to maintain synapsis of DNA ends than by sealing one strand. The unique characterisgc (among vertebrate DNA ligases) of ligase 4 as a single-turnover enzyme (51) is curious, and obviously infers that the ligase 4 molecule that resolves the first strand break does not (necessarily) resolve the second strand break (which could be recognized as a nick or single-stranded DNA gap). This may reflect LX4’s important structural role both in recruigng other enzymes, and in promogng stable synapsis. As noted above, emerging studies demonstrate not only that Lig3 can associate with NHEJ factors, but also that when LX4 is inacgvated by mutagon, Lig3 clearly funcgons to repair DSBs, in an inacgve LX4-dependent manner, in living cells and animals (10, 14). 8 Figure 2: Numerous NHEJ complexes have been visualized by cryo-EM. Cartoons depicgng NHEJ long-range complexes (Ku80-medieated and XLF-dependent dimer), long-range ATP (LR-ATP) complex, DNA-PKcs homodimer, and SRCs. DNA-PKcs is colored pink, Ku-green, XLF-red, XRCC4- blue, Lig4 (fuscia) (derived from PDB 6ZHE, PDB 7NFC, PDF 7LT3, PDB 8EZB, PDB 8BYH, PDB 7LSY, PDB 8EZ9). Cryo-EM structures of NHEJ synapZc complexes. This two-stage synapgc model of NHEJ has been strengthened by recent structural work describing Cryo-EM structures of mulgmeric NHEJ complexes posigoned around breaks. The published structures include descripgons of two major forms of NHEJ complexes that synapse DNA ends ~115Å apart (consistent with long-range synapgc complexes). These dimers are organized around two disgnct interfaces: one mediated by a trans interacgon between DNA-PKcs and Ku80’s extreme C-terminus, and the other dependent 9 on a centrally placed XLF homodimer interacgng with Ku and the LX4 complex, which also involves a large interface between the two DNA-PKcs molecules (Figure 2). These dimers have been termed the Ku80-mediated dimer (or domain-swap dimer), and the XLF-dependent dimer. Inigal observagons of the Ku80-mediated dimer only included the components of the DNA-PK complex, which would be consistent with the LRC requiring only Ku and DNA-PKcs. However, further studies show that the Ku80-mediated dimer can also contain XLF, PAXX, and LX4, or XLF and LX4, or PAXX and LX4 (22). The XLF-dependent dimer was first observed with two DNA-PK protomers, two LX4 complexes and one XLF homodimer. Recent studies have shown that PAXX can also assemble into this complex (22). Figure 2 presents cartoon depicgons of the dimers (for simplicity, the complexes that include PAXX are not represented); structural models of these complexes are available in recent excellent reviews (52, 53) He and colleagues have shown that in the presence of ATP, the XLF-dependent dimer can progress to a new form, termed long-range ATP (LR-ATP) (54). In this complex, ABCDE phosphorylagon has occurred (likely in trans) resulgng in a major conformagonal change: each DNA-PK protomer has a remarkably similar structure to the ABCDE phosphorylated monomeric complex with the DEB dissolved, and the phosphorylated ABCDE sites anchored by K/R residues in M-HEAT. These authors also observed a DNA-PKcs homodimer that shares some structural similarity with homodimers of other PIKKs, ATM and ATR. They hypothesized that this dimer formed amer ATP-induced dissociagon of DNA-PKcs from Ku/DNA. Another idea proposed was that this homodimer might serve as a DNA-PKcs reservoir. Notably, although these DNA-PKcs dimers were observed without the other components of the XLF-dependent dimer, no complexes consistent with SRC were observed in the cryo-EM experiments with ATP. Cryo-EM structures of NHEJ complexes consistent with SRCs have only been obtained in experiments uglizing Ku, LX4, and XLF with a DNA substrate that facilitates end synapsis (18). The organizagon of this short- range synapgc complex is remarkably like the organizagon observed in the XLF-dependent dimer, with a single XLF monomer mediagng the synapsis between two Ku-bound DNA ends, and LX4/XLF/LX4 bridge promogng synapsis (Figure 2). Of note, in the short-range structure, although the catalygc domain of only one LX4 is structured and posigoned over the perfectly juxtaposed DNA ends, the second LX4 complex is clearly present, which is not consistent with the 10 stoichiometry discussed above. Sgnson et al. suggest that one LX4 complex is released at the gme of long-range to short-range transigon but noted that another LX4 may associate (48) (thus, two potengal SRCs are depicted in Figure 2). Finally, a DNA-PK trimer was observed (not shown), consisgng of a central DNA-PK protomer, interacgng with another to form an XLF-dependent dimer, and concurrently interacgng with a third DNA-PK protomer via the Ku80-mediated domain swap interacgon. It is not engrely clear how this trimeric structure would funcgon, but at least two disgnct DSBs would be required (55). AlternaZve models of NHEJ synapsis. The observagon that DNA-PKcs catalygc acgvity is required for SRC formagon in the X. Laevis extract model has generated substangal discussion over the role of DNA-PKcs when presengng complete models for NHEJ synapsis. Although the idea that DNA-PK facilitates synapsis was proposed decades ago, originally by Chu and colleagues (56) and supported by others (57), in other elegant smFRET studies using purified human NHEJ components, stable synapsis of DNA ends is not dependent on DNA-PKcs6 (58–60) Sgll, other experimental systems using purified human proteins, demonstrate a clear role for DNA-PKcs in synapsis (61). In the studies from Lieber and colleagues (58–60), adding DNA-PKcs decreased the total number of synapgc foci. These invesggators also examined the role of DNA end chemistry on LX4 mediated synapsis and ligagon; their studies showed a strong dependence for 5’phosphates for synapsis in the absence of DNA-PKcs (59). A caveat of this and other smFRET approaches is the inability to differengate between synapsis-mediated or covalent ligagon of the two oligonucleogdes. Further in vitro single-molecule work (58) provided evidence for a long- range synapgc complex formed from purified human Ku70/80 and LX4. Addigon of purified XLF to these reacgons induced a transigon into a short-range synapgc complex, a consistent observagon with their earlier findings of DNA-PKcs-independent short-range complex formagon. Thus, addigonal work will be required to elucidate how disgnct NHEJ complexes facilitate repair. Sgll, it seems likely that there are complex, cell-wide effects of losing DNA-PKcs beyond its role in synapsing and regulagng access to DNA ends in NHEJ. There is ample evidence for a mulgfaceted role for DNA-PKcs in both directly synapsing ends and pargcipagng in the cell-wide DNA damage response; moreover, DNA-PK has been implicated to funcgon in numerous other cellular funcgons [reviewed in (62)]. One argument suggesgng that DNA-PKcs can be dispensable 11 for NHEJ is that cells lacking DNA-PKcs are notably less sensigve to DSB inducing drugs than isogenic cells lacking other core NHEJ factors (63, 64). However, this conclusion is complicated by the fact that cells lacking DNA-PKcs also lose significant expression of ATM, leading to an impaired apoptogc response (65, 66). In fact, loss of even one allele of ATM substangally rescues the severe phenotype of Lig4 deficient mice (67). There are also obvious examples of DNA-PKcs-independent NHEJ. Although recent phylogenegc work from Lees-Miller et al. have established that DNA-PKcs is highly represented in many species far removed from vertebrates (35), there are sgll many species with funcgonal NHEJ pathways that lack DNA-PKcs homologues (for example, some insects, yeast, and bacteria) (68). Addigonally, the recently published cryo-EM structure of the putagve short-range complex was observed in cryo-EM experiments that lacked DNA-PKcs; thus, short-range complex assembly can occur in the absence of DNA-PKcs (18). Moreover, because of the iteragve nature of NHEJ, we must consider that there may be mulgple paths that lead to the same outcome: there is more than one way to ge a knot/ligate an end. In sum, while recent studies and technological advancements have provided new insights, substangal gaps in knowledge remain that must be resolved before a full understanding of how NHEJ funcgons in living cells to rapidly repair DSB while minimizing genome alteragon. Many, but not all NHEJ end-processing steps occur in short-range complexes. Another transformagve study from Loparo and colleagues (21) provided compelling biochemical evidence that factors required for the formagon of SRCs [Ku, XLF, LX4, and the catalygc acgvity of DNA-PK] are also essengal to enable fill-in synthesis mediated by X-family polymerases [polƛ and polµ, 3’ adduct cleavage mediated by Tdp1, phosphorylagon of 5’ hydroxyls by PNKP, and the acgvity of an unidengfied 5’>3’ exonuclease on 5’ flaps (21). Moreover, DNA ends were observed to be protected from end-processing acgviges in the LRC. At first, these results might seem at odds with our conclusion that the two long-range complexes promote different aspects of end-processing; this is not the case. The mutagonal studies suggest that the SRC derives from the XLF-mediated dimer; the enzymagc acgviges absolutely ascribed to the SRC by Sgnson et al., (except for the unidengfied 5'>3" nuclease) should all promote fill-in end processing. So, the XLF-dependent dimer promotes fill-in end 12 processing by promogng progression to the SRC. Part of the argument from Sgnson et al that nuclease acgvity is limited to the SRC was that DNA-PK inhibigon blocked nuclease acgvity, presumably because this blocks progression to the SRC. However, DNA-PK inhibigon also blocks the ABCDE phosphorylated form of DNA-PK acgvagon that is requisite to Artemis acgvagon and potengally required for the acgvagon of other nucleases that may funcgon in NHEJ (Mre11, Apollo)(1, 69). Thus, what is less clear is what nucleases (besides Artemis) contribute to NHEJ and in what NHEJ complexes do these nucleases funcgon. This concludes the work adapted from DNA-PK: A synopsis beyond synapsis. L4’s role in dimeric NHEJ complexes: In recent NHEJ cryo-EM structures, the XRCC4-L4 complex has been observed in variagons of both classes of NHEJ dimers, with the catalygc core only observed in the SRC and one unpublished version of the Ku80-mediated LRC (4–6). The LX4 complex is seen to interact with Ku70 and Ku80 through XRCC4’s head domain and L4’s BRCT repeats (Figure 1, 3) (18, 19, 22, 71). In the XLF-mediated dimer, LX4 instead facilitates an interacgon with an XLF homodimer spanning the gap between ends, bridging each Ku “donut” (there is also a conformagon change in DNA-PKcs to more gghtly bind ends with a trans PKcs- PKcs interface supporgng synapsis in addigon to LX4-XLF-LX4, Figure 3). The Ku80-mediated dimer was inigally observed in the absence of LX4, however recent studies from Chaplin and colleagues incorporagng PAXX and LX4 have since resolved a version of the Ku80-mediated dimer with LX4 bound to DNA-PK, but not supporgng the synapse (in contrast to their role in supporgng the XLF-mediated dimer). The short-range complex structure contains two LX4 complexes, with one interacgng only though its LX4 structural domain and the other directly bound to the break (Figure 4)(18)—notably contradicgng recent smFRET work from Loparo and colleagues (48) that indicates one molecule of L4 leaves the break prior to SR synapsis. 13 Figure 3: XLF-Mediated Long-Range Complex. PDB 7LSY: a second dimeric structure of DNA- PKcs (grey), Ku70/Ku80 (grey/black), L4 (green), XRCC4 (purple), and XLF (orange), work together to synapse DNA (orange) in a long-range synapse. Here, LX4 synapses ends through interacgons with Ku80 and XLF, along with a DNA-PKcs trans interacgon in the head domain. 14 Figure 4: DNA Ligase IV plays a pivotal role in the observed CryoEM Complex. PDB 7LSY: Ku70/Ku80 (Black), L4 (Green), XRCC4 (Purple), XLF (Orange) work together to synapse DNA (yellow) in a ligagon-competent state. This structure features two LX4 complexes, with the rightmost L4 catalygc core bound to the DNA end. Only the structural domains (L4 BRCT, XRCC4 homodimer) are observed for the second LX4 complex (lem). Diverse DNA substrates and the cell cycle dictate repair pathway choice: While it’s somewhat unconvengonal to think of DNA repair enzymes in terms of classical enzyme kinegcs—since the concentragon of a repair factor typically dwarfs the concentragon of DSBs—the model works well for considering DNA end chemistry as a key element in pathway choice. The diverse mechanisms that generate DSBs results in a very wide range of substrates—ranging from unique end adducts formed by many chemotherapeugc agents, secondary structures biased near the break, single- ended DSBs formed at failed replicagon forks, to ends compromised by previous repair steps. To this end, eukaryogc organisms have evolved several mechanisms to sense breaks and engage the correct repair mechanism depending on substrate chemistry and cell cycle status. Homologous recombinaZon: The other major DSB repair pathway, homologous recombinagon (HR), serves as a slow, energy intensive, high-fidelity repair pathway that can repair nearly every variagon of DSB chemistry (with the notable excepgon of programmed DSBs generated in 15 VDJ/CSR recombinagon). The primary drawback to HR is gming: namely, it’s only available in the S/G2 phases of the cell cycle amer DNA replicagon generates a nearby homologous template for repair. This limits repair to acgvely dividing cells that encounter breaks behind replicagon forks. Furthermore, instead of directly repairing ends, HR first resects large stretches of DNA to generate long single-stranded ends. This resecgon (mediated by MRN and Exo1) serves to clear lesions and facilitate homology search prior to synthesis of new DNA—thereby creagng a “high fidelity” repaired region (assuming the homologous template matches the original sequence). The resecgon and homology search are a gme-intensive processes, requiring mulgple rounds of processing (including resolugon of the DNA tetramer holiday juncgons) prior to finalizing repair. The intense processing of ends directed to HR results in intermediates incompagble with NHEJ; as such, the pathways act compeggvely and there are mulgple mechanisms to restrain either pathway depending on context. These mechanisms act 1) to restrain NHEJ and prevent premature ligagon (possibly by prevengng transigon into the SRC)(21) and 2) acgvely remove NHEJ factors from acgng on ends that are targeted towards HR (3, 72). Interesgngly, a recent live- cell SMI study indicates the same aborgve mechanism engages in cells lacking L4 (73)—implying this mechanism may more broadly engage if NHEJ is incapable of rapidly transigoning ends into the SRC. AlternaZve end joining: (alt-EJ) serves as less well-defined collecgon of factors that’s available to join breaks not resolved by either of the “canonical” NHEJ or HR pathways. While the pathway is loosely defined, recent studies (including experiments reported in Chapter 3) suggest that DNA Polymerase Theta (Polθ) is a crigcal factor essengal for many alt-EJ repair events. Polθ is a large and uniquely contains both a highly error-prone polymerase domain and a helicase domain capable of bridging ends uglizing 1-5 bp of terminal microhomology (74). The dual helicase- polymerase funcgon is generally thought to facilitate synthesis through extremely difficult to replicate stretches of DNA (g quadruplexes, other complicated secondary structures) in addigon to its role as a backup end-joining factor (74–76). Of note, Polθ prefers free DNA end substrates with long secgons of 3’ ssDNA (77, 78) and its end-joining funcgons likely support joining of failed HR intermediates (where resecgon can produce long single-stranded ends that are poor substrates for canonical NHEJ). While further 16 work needs to be done, current literature supports MMEJ—and its dependence on Polθ helicase/polymerase—as a key mediator of joining in the absence of NHEJ. Alt-EJ serves as an interesgng foil to NHEJ, being both lower fidelity and significantly slower. These qualiges reflect alt-EJ’s typical substrates: rare complex breaks that “fail” out of the two major DSB repair pathways. As such, there is lijle evolugonary pressure for alt-EJ to have developed mechanisms to respond to many breaks, quickly. This is in stark contrast to NHEJ, where factors have evolved to be both fast, highly regulated, and relagvely accurate. It may be more accurate to characterize alt-EJ as a collecgon of backup mechanisms that may really be secondary funcgons of many other DNA repair pathways working together to resolve DSBs. Many factors hypothesized to resolve breaks in the absence of NHEJ have notable roles in other pathways. For example, there is sgll some residual end joining funcgon in the absence of L4, but neither Ligase I nor Ligase III becomes essengal following loss of L4 (nuclear loss of both L1 and L3 is synthegcally lethal regardless of L4 status). Furthermore, the slower kinegcs of MMEJ indicate that it is not likely in direct compeggon to NHEJ or HR (which do act in direct compeggon), and instead works as a “catch-all” mechanism that only acts if higher-fidelity pathways are unable to resolve a pargcular break in a gmely manner. CONCLUSIONS I would like to leave this chapter highlighgng the remarkable advancements in our understanding of NHEJ over the course of my PhD. When I began, there was significant disagreement over DNA synapsis models—with arguments over the significance of DNA-PKcs vs XRCC4/XLF filaments as a primary synapgc mechanism. The applicagon of clever SMI experiments to address NHEJ factor kinegcs and dedicagon of Cryo-EM groups to generate 3D-maps of NHEJ complexes has truly deepened the field’s appreciagon for how flexible the pathway can be. I look at this project through the lens of those advancements. From March 1st, 2020, I’ve sought to understand L4’s structural role in supporgng NHEJ—before we understood anything about the disgnct LR complexes or true organizagon of the SR complex. I’ve since focused my PhD work— including much work that is not included in this dissertagon—through the lens of structure- funcgon relagonships between NHEJ’s structural factors (XRCC4, XLF, Ku70/80) and L4, advancing our understanding the structural role of L4 and how it promotes end joining beyond catalysis. 17 1. 2. 3. 4. 5. 6. 7. 8. 9. REFERENCES Sonmez,C., Toia,B., Eickhoff,P., Matei,A.M., El Beyrouthy,M., Wallner,B., Douglas,M.E., de Lange,T. and Lojersberger,F. (2024) DNA-PK controls Apollo’s access to leading-end telomeres. Nucleic Acids Research, 10.1093/nar/gkae105. Wang,Y., Ghosh,G. and Hendrickson,E.A. (2009) Ku86 represses lethal telomere delegon events in human somagc cells. Proceedings of the Na9onal Academy of Sciences, 106, 12430–12435. Bossaert,M., Moreno,A.T., Peixoto,A., Pillaire,M.-J., Chanut,P., Frit,P., Calsou,P., Loparo,J.J. and Brijon,S. (2024) Idengficagon of the main barriers to Ku accumulagon in chromagn. Cell Reports, 43. Kragelund,B.B., Weterings,E., Hartmann-Petersen,R. and Keijzers,G. (2016) The Ku70/80 ring in Non-Homologous End-Joining: easy to slip on, hard to remove. Front Biosci (Landmark Ed), 21, 514–527. Weterings,E., Verkaik,N.S., Keijzers,G., Florea,B.I., Wang,S.-Y., Ortega,L.G., Uematsu,N., Chen,D.J. and van Gent,D.C. (2009) The Ku80 Carboxy Terminus Sgmulates Joining and Artemis-Mediated Processing of DNA Ends. MCB, 29, 1134–1142. Inagawa,T., Wennink,T., Lebbink,J., Keijzers,G., Florea,B., Verkaik,N. and van Gent,D. (2020) C-Terminal Extensions of Ku70 and Ku80 Differengally Influence DNA End Binding Properges. INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, 21. Singleton,B.K., Torres-Arzayus,M.I., Ro~nghaus,S.T., Taccioli,G.E. and Jeggo,P.A. (1999) The C Terminus of Ku80 Acgvates the DNA-Dependent Protein Kinase Catalygc Subunit. Mol. Cell. Biol., 19, 3267–3277. Sibanda,B.L., Critchlow,S.E., Begun,J., Pei,X.Y., Jackson,S.P., Blundell,T.L. and Pellegrini,L. (2001) Crystal structure of an Xrcc4–DNA ligase IV complex. Nat Struct Mol Biol, 8, 1015– 1019. Grawunder,U., Zimmer,D. and Lieber,M.R. (1998) DNA ligase IV binds to XRCC4 via a mogf located between rather than within its BRCT domains. Current Biology, 8, 873–879. 10. Goff,N.J., Brenière,M., Buehl,C.J., de Melo,A.J., Huskova,H., Ochi,T., Blundell,T.L., Mao,W., Yu,K., Modesg,M., et al. (2022) Catalygcally inacgve DNA ligase IV promotes DNA repair in living cells. Nucleic Acids Research, 10.1093/nar/gkac913. 11. Meek,K. (2020) Acgvagon of DNA-PK by hairpinned DNA ends reveals a stepwise mechanism of kinase acgvagon. Nucleic Acids Research, 48, 9098–9108. 12. Roy,S., de Melo,A., Xu,Y., Tadi,S., Negrel,A., Hendrickson,E., Modesg,M. and Meek,K. (2015) XRCC4/XLF Interacgon Is Variably Required for DNA Repair and Is Not Required for Ligase IV Sgmulagon. MOLECULAR AND CELLULAR BIOLOGY, 35, 3017–3028. 18 13. Yu,Y., Wang,W., Ding,Q., Ye,R., Chen,D., Merkle,D., Schriemer,D., Meek,K. and Lees- Miller,S. (2003) DNA-PK phosphorylagon sites in XRCC4 are not required for survival amer radiagon or for V(D)J recombinagon. DNA REPAIR, 2, 1239–1252. 14. Medina-Suarez,D., Han,L., O’Reily,S., Liu,J., Wei,C., Brenière,M., Goff,N.J., Chen,C., Modesg,M., Meek,K., et al. (2024) DNA ligases 3 and 4 cooperate in vivo to facilitate DNA repair and organism viability. In Review. 15. Masani,S., Han,L., Meek,K. and Yu,K. (2016) Redundant funcgon of DNA ligase 1 and 3 in alternagve end-joining during immunoglobulin class switch recombinagon. PNAS, 113, 1261–1266. 16. Oh,S., Harvey,A., Zimbric,J., Wang,Y., Nguyen,T., Jackson,P.J. and Hendrickson,E.A. (2014) DNA ligase III and DNA ligase IV carry out genegcally disgnct forms of end joining in human somagc cells. DNA Repair, 21, 97–110. 17. 18. 19. 20. 21. 22. 23. Vu,D.-D., Bonucci,A., Brenière,M., Cisneros-Aguirre,M., Pelupessy,P., Wang,Z., Carlier,L., Bouvignies,G., Cortes,P., Aggarwal,A.K., et al. (2024) Mulgvalent interacgons of the disordered regions of XLF and XRCC4 foster robust cellular NHEJ and drive the formagon of ligagon-boosgng condensates in vitro. Nat Struct Mol Biol, 10.1038/s41594-024- 01339-x. Chen,S., Lee,L., Naila,T., Fishbain,S., Wang,A., Tomkinson,A.E., Lees-Miller,S.P. and He,Y. (2021) Structural basis of long-range to short-range synapgc transigon in NHEJ. Nature, 10.1038/s41586-021-03458-7. Chaplin,A.K., Hardwick,S.W., Stavridi,A.K., Buehl,C.J., Goff,N.J., Ropars,V., Liang,S., Oliveira,T.M.D., Chirgadze,D.Y., Meek,K., et al. (2021) Cryo-EM of NHEJ supercomplexes provides insights into DNA repair. Molecular Cell, 0. Buehl,C.J., Goff,N.J., Hardwick,S.W., Gellert,M., Blundell,T.L., Yang,W., Chaplin,A.K. and Meek,K. (2023) Two disgnct long-range synapgc complexes promote different aspects of end processing prior to repair of DNA breaks by non-homologous end joining. Molecular Cell, 10.1016/j.molcel.2023.01.012. Sgnson,B.M., Moreno,A.T., Walter,J.C. and Loparo,J.J. (2020) A Mechanism to Minimize Errors during Non-homologous End Joining. Molecular Cell, 77, 1080-1091.e8. Seif-El-Dahan,M., Kefala-Stavridi,A., Frit,P., Hardwick,S.W., Chirgadze,D.Y., Maia De Oliviera,T., Brijon,S., Barboule,N., Bossaert,M., Pandurangan,A.P., et al. (2023) PAXX binding to the NHEJ machinery explains funcgonal redundancy with XLF. Science Advances, 9, eadg2834. Tadi,S., Tellier-Lebegue,C., Nemoz,C., Drevet,P., Audebert,S., Roy,S., Meek,K., Charbonnier,J. and Modesg,M. (2016) PAXX Is an Accessory c-NHEJ Factor that Associates with Ku70 and Has Overlapping Funcgons with XLF. CELL REPORTS, 17, 541–555. 19 24. 25. 26. 27. 28. Roy,S., Andres,S., Vergnes,A., Neal,J., Xu,Y., Yu,Y., Lees-Miller,S., Junop,M., Modesg,M. and Meek,K. (2012) XRCC4’s interacgon with XLF is required for coding (but not signal) end joining. NUCLEIC ACIDS RESEARCH, 40, 1684–1694. Balmus,G., Barros,A.C., Wijnhoven,P.W.G., Lescale,C., Hasse,H.L., Boroviak,K., Sage,C. le, Doe,B., Speak,A.O., Galli,A., et al. (2016) Synthegc lethality between PAXX and XLF in mammalian development. Genes Dev., 30, 2152–2157. Cisneros-Aguirre,M., Lopezcolorado,F.W., Tsai,L.J., Bhargava,R. and Stark,J.M. (2022) The importance of DNAPKcs for blunt DNA end joining is magnified when XLF is weakened. Nat Commun, 13, 3662. Graham,T.G.W., Carney,S.M., Walter,J.C. and Loparo,J.J. (2018) A single XLF dimer bridges DNA ends during nonhomologous end joining. Nat Struct Mol Biol, 25, 877–884. Bhargava,R., Sandhu,M., Muk,S., Lee,G., Vaidehi,N. and Stark,J.M. (2018) C-NHEJ without indels is robust and requires synergisgc funcgon of disgnct XLF domains. Nat Commun, 9, 2484. 29. Ochi,T. and Khan,H. (2023) Plant PAXX has an XLF-like funcgon and sgmulates DNA end- joining by the Ku-DNA ligase IV-XRCC4 complex. 10.1101/2023.05.26.542399. 30. Yu,Y., Gupta,S., (2005) Cui,X., Autophosphorylagon of DNA-Dependent Protein Kinase Regulates DNA End Processing and May Also Alter Double-Strand Break Repair Pathway Choice. MCB, 25, 10842–10852. Lees-Miller,S.P. and Meek,K. Cho,Y.-M., 31. Neal,J.A., Sugiman-Marangos,S., VanderVere-Carozza,P., Wagner,M., Turchi,J., Lees- Miller,S.P., Junop,M.S. and Meek,K. (2014) Unraveling the Complexiges of DNA- Dependent Protein Kinase Autophosphorylagon. Molecular and Cellular Biology, 34, 2162–2175. 32. Liu,L., Chen,X., Li,J., Wang,H., Buehl,C.J., Goff,N.J., Meek,K., Yang,W. and Gellert,M. (2021) Autophosphorylagon transforms DNA-PK from protecgng to processing DNA ends. Molecular Cell, 0. 33. Öz,R., Wang,J.L., Guerois,R., Goyal,G., KK,S., Ropars,V., Sharma,R., Koca,F., Charbonnier,J.-B., Modesg,M., et al. (2021) Dynamics of Ku and bacterial non- homologous end-joining characterized using single DNA molecule analysis. Nucleic Acids Research, 49, 2629–2641. 34. Manoj,F. and Kuhlman,T.E. (2023) Phylogenegc Distribugon of Prokaryogc Non- in Bacteria and Archaea. Homologous End 10.1101/2023.09.30.560322. Joining DNA Repair Systems 35. Lees-Miller,J.P., Cobban,A., Katsonis,P., Bacolla,A., Tsutakawa,S.E., Hammel,M., Meek,K., Anderson,D.W., Lichtarge,O., Tainer,J.A., et al. (2021) Uncovering DNA-PKcs ancient 20 phylogeny, unique sequence mogfs and insights for human disease. Progress in Biophysics and Molecular Biology, 163, 87–108. Lakshmipathy,U. and Campbell,C. (1999) The Human DNA Ligase III Gene Encodes Nuclear and Mitochondrial Proteins. Molecular and Cellular Biology, 19, 3869. Ellenberger,T. and Tomkinson,A.E. (2008) Eukaryogc DNA Ligases: Structural and Funcgonal Insights. Annual Review of Biochemistry, 77, 313–338. Sallmyr,A., Khajri Bhandari,S., Naila,T. and Tomkinson,A.E. (2023) Mammalian DNA ligases; roles integrity. Journal of Molecular Biology, 10.1016/j.jmb.2023.168276. in maintaining genome Sallmyr,A., Rashid,I., Bhandari,S.K., Naila,T. and Tomkinson,A.E. (2020) Human DNA ligases in replicagon and repair. DNA Repair, 93, 102908. Doherty,A.J. and Suh,S.W. (2000) Structural and mechanisgc conservagon in DNA ligases. Nucleic Acids Research, 28, 4051–4058. Shuman,S. (2009) DNA Ligases: Progress and Prospects*. Journal of Biological Chemistry, 284, 17365–17369. Conlin,M.P., Reid,D.A., Small,G.W., Chang,H.H., Watanabe,G., Lieber,M.R., Ramsden,D.A. IV Guides End-Processing Choice during and Rothenberg,E. (2017) DNA Ligase Nonhomologous End Joining. Cell Reports, 20, 2810–2819. Chisgakov,D.A., Voronova,N.V. and Chisgakov,A.P. (2009) Ligase IV syndrome. European Journal of Medical Gene9cs, 52, 373–378. Han,L., Masani,S., Hsieh,C. and Yu,K. (2014) DNA Ligase I Is Not Essengal for Mammalian Cell Viability. Cell Reports, 7, 316–320. Goff,N.J., Mikhova,M., Schmidt,J.C. and Meek,K. (2024) DNA-PK: A synopsis beyond synapsis. DNA Repair, 141, 103716. Graham,T.G.W., Walter,J.C. and Loparo,J.J. (2016) Two-Stage Synapsis of DNA Ends during Non-homologous End Joining. Molecular Cell, 61, 850–858. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. Meek,K., Lees-Miller,S. and Modesg,M. (2012) N-terminal constraint acgvates the catalygc subunit of the DNA-dependent protein kinase in the absence of DNA or Ku. NUCLEIC ACIDS RESEARCH, 40, 2964–2973. 48. Sgnson,B.M., Carney,S.M., Walter,J.C. and Loparo,J.J. (2024) Structural role for DNA Ligase IV in promogng the fidelity of non-homologous end joining. Nat Commun, 15, 1250. 49. Waters,C.A., Strande,N.T., Pryor,J.M., Strom,C.N., Mieczkowski,P., Burkhalter,M.D., Oh,S., Qaqish,B.F., Moore,D.T., Hendrickson,E.A., et al. (2014) The fidelity of the ligagon 21 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. step determines how ends are resolved during nonhomologous end joining. Nat Commun, 5, 4286. Gu,J., Lu,H., Tippin,B., Shimazaki,N., Goodman,M.F. and Lieber,M.R. (2007) XRCC4:DNA ligase IV can ligate incompagble DNA ends and can ligate across gaps. The EMBO Journal, 26, 1010–1023. Chen,X., Ballin,J.D., Della-Maria,J., Tsai,M.-S., White,E.J., Tomkinson,A.E. and Wilson,G.M. (2009) Disgnct kinegcs of human DNA ligases I, IIIα, IIIβ, and IV reveal direct DNA sensing ability and differengal physiological funcgons in DNA repair. DNA Repair, 8, 961–968. Amin,H., Zahid,S., Hall,C. and Chaplin,A.K. (2024) Cold snapshots of DNA repair: Cryo-EM structures of DNA-PKcs and NHEJ machinery. Progress in Biophysics and Molecular Biology, 186, 1–13. Vogt,A., He,Y. and Lees-Miller,S.P. (2023) How to fix DNA breaks: new insights into the mechanism of non-homologous end joining. Biochemical Society Transac9ons, 10.1042/BST20220741. Chen,S., Vogt,A., Lee,L., Naila,T., McKeown,R., Tomkinson,A.E., Lees-Miller,S.P. and He,Y. (2023) Cryo-EM visualizagon of DNA-PKcs structural intermediates in NHEJ. Science Advances, 9, eadg2838. Hardwick,S.W., Stavridi,A.K., Chirgadze,D.Y., De Oliveira,T.M., Charbonnier,J.-B., Ropars,V., Meek,K., Blundell,T.L. and Chaplin,A.K. (2023) Cryo-EM structure of a DNA-PK trimer: higher order oligomerisagon in NHEJ. Structure, 31, 895-902.e3. DeFazio,L.G., Stansel,R.M., Griffith,J.D. and Chu,G. (2002) Synapsis of DNA ends by DNA- dependent protein kinase. The EMBO journal, 21, 3192–3200. Hammel,M., Yu,Y., Mahaney,B.L., Cai,B., Ye,R., Phipps,B.M., Rambo,R.P., Hura,G.L., Pelikan,M., So,S., et al. (2010) Ku and DNA-dependent protein kinase dynamic conformagons and assembly regulate DNA binding and the inigal non-homologous end joining complex. The Journal of biological chemistry, 285, 1414–1423. Zhao,B., Watanabe,G., Morten,M.J., Reid,D.A., Rothenberg,E. and Lieber,M.R. (2019) The essengal elements for the noncovalent associagon of two DNA ends during NHEJ synapsis. Nat Commun, 10, 3588. Reid,D.A., Conlin,M.P., Yin,Y., Chang,H.H., Watanabe,G., Lieber,M.R., Ramsden,D.A. and Rothenberg,E. (2017) Bridging of double-stranded breaks by the nonhomologous end- joining ligagon complex is modulated by DNA end chemistry. Nucleic Acids Research, 45, 1872–1878. 60. Reid,D.A., Keegan,S., Leo-Macias,A., Watanabe,G., Strande,N.T., Chang,H.H., Oksuz,B.A., Fenyo,D., Lieber,M.R., Ramsden,D.A., et al. (2015) Organizagon and dynamics of the 22 nonhomologous end-joining machinery during DNA double-strand break repair. Proceedings of the Na9onal Academy of Sciences, 112, E2575–E2584. 61. Wang,J.L., Duboc,C., Wu,Q., Ochi,T., Liang,S., Tsutakawa,S.E., Lees-Miller,S.P., Nadal,M., Tainer,J.A., Blundell,T.L., et al. (2018) Dissecgon of DNA double-strand-break repair using novel single-molecule forceps. Nature structural & molecular biology, 25, 482–487. 62. 63. 64. 65. Dylgjeri,E., McNair,C., Goodwin,J.F., Raymon,H.K., McCue,P.A., Shafi,A.A., Leiby,B.E., Leeuw,R., Kothari,V., McCann,J.J., et al. (2019) Pleiotropic impact of DNA-PK in cancer and implicagons for therapeugc strategies. Clinical cancer research : an official journal of the American Associa9on for Cancer Research, 25, 5623–5637. Beucher,A., Birraux,J., Tchouandong,L., Barton,O., Shibata,A., Conrad,S., Goodarzi,A.A., Krempler,A., Jeggo,P.A. and Lobrich,M. (2009) ATM and Artemis promote homologous recombinagon of radiagon-induced DNA double-strand breaks in G2. The EMBO journal, 28, 3413–3427. Rooney,S., Alt,F.W., Lombard,D., Whitlow,S., Eckersdorff,M., Fleming,J., Fugmann,S., Ferguson,D.O., Schatz,D.G. and Sekiguchi,J. (2003) Defecgve DNA repair and increased genomic instability in Artemis-deficient murine cells. The Journal of experimental medicine, 197, 553–565. Peng,Y., Woods,R.G., Beamish,H., Ye,R., Lees-Miller,S.P., Lavin,M.F. and Bedford,J.S. (2005) Deficiency in the catalygc subunit of DNA-dependent protein kinase causes down- regulagon of ATM. Cancer Res, 65, 1670–1677. 66. Neal,J. and Meek,K. (2019) Deciphering phenotypic variance in different models of DNA- PKcs deficiency. DNA REPAIR, 73, 7–16. 67. 68. 69. 70. Sekiguchi,J., Ferguson,D.O., Chen,H.T., Yang,E.M., Earle,J., Frank,K., Whitlow,S., Gu,Y., Xu,Y., Nussenzweig,A., et al. (2001) Genegc interacgons between ATM and the nonhomologous end-joining factors in genomic stability and development. In Proceedings of the na9onal academy of sciences of the united states of america.Vol. 98, pp. 3243– 3248. Bertrand,C., Thibessard,A., Bruand,C., Lecointe,F. and Leblond,P. (2019) Bacterial NHEJ: a never ending story. Mol Microbiol, 111, 1139–1151. Deshpande,R.A., Myler,L.R., Soniat,M.M., Makharashvili,N., Lee,L., Lees-Miller,S.P., Finkelstein,I.J. and Paull,T.T. (2020) DNA-dependent protein kinase promotes DNA end processing by MRN and CtIP. Sci Adv, 6, 0922. Chaplin,A.K., Hardwick,S.W., Liang,S., Kefala Stavridi,A., Hnizda,A., Cooper,L.R., De Oliveira,T.M., Chirgadze,D.Y. and Blundell,T.L. (2021) Dimers of DNA-PK create a stage for DNA double-strand break repair. Nat Struct Mol Biol, 28, 13–19. 23 71. 72. Chen,X., Xu,X., Chen,Y., Cheung,J.C., Wang,H., Jiang,J., de Val,N., Fox,T., Gellert,M. and Yang,W. (2021) Structure of an acgvated DNA-PK and its implicagons for NHEJ. Molecular Cell, 81, 801-810.e3. Balmus,G., Pilger,D., Coates,J., Demir,M., Sczaniecka-Clim,M., Barros,A.C., Woods,M., Fu,B., Yang,F., Chen,E., et al. (2019) ATM orchestrates the DNA-damage response to counter toxic non-homologous end-joining at broken replicagon forks. Nat Commun, 10, 87. 73. Mikhova,M., Goff,N.J., Janovič,T., Heyza,J.R., Meek,K. and Schmidt,J.C. (2024) Single- molecule imaging reveals the kinegcs of non-homologous end-joining in living cells. 10.1101/2023.06.22.546088. 74. 75. 76. Schaub,J.M., Soniat,M.M. and Finkelstein,I.J. (2022) Polymerase theta-helicase promotes end joining by stripping single-stranded DNA-binding proteins and bridging DNA ends. Nucleic Acids Research, 10.1093/nar/gkac119. Beagan,K. and McVey,M. (2016) Linking DNA polymerase theta structure and funcgon in health and disease. Cell. Mol. Life Sci., 73, 603–615. Li,C., Zhu,H., Jin,S., Maksoud,L.M., Jain,N., Sun,J. and Gao,Y. (2023) Structural basis of DNA polymerase θ mediated DNA end joining. Nucleic Acids Research, 51, 463–474. 77. Wyaj,D.W., Feng,W., Conlin,M.P., Yousefzadeh,M.J., Roberts,S.A., Mieczkowski,P., Wood,R.D., Gupta,G.P. and Ramsden,D.A. (2016) Essengal Roles for Polymerase theta- Mediated End Joining in the Repair of Chromosome Breaks. Mol. Cell, 63, 662–673. 78. He,P. and Yang,W. (2018) Template and primer requirements for DNA Pol θ-mediated end joining. Proceedings of the Na9onal Academy of Sciences, 115, 7747–7752. 24 Chapter 2: Catalytically inactive DNA ligase IV promotes DNA repair in living cells. By: Noah J. Goff1,3,4, Manon Brenière5, Christopher J. Buehl1,3,4, Abinadabe J. de Melo5, Hana Huskova5, Takashi Ochi6, Tom Blundell7, Weifeng Mao2,3,8, Kefei Yu2,3, Mauro Modesg5,, and Katheryn Meek1,3,4*. Data chapter 1College of Veterinary Medicine, 2College of Human Medicine, 3Department of Microbiology & Molecular Gene;cs, 4Department of Pathobiology & Diagnos;c Inves;ga;on, Michigan State University, East Lansing, MI 48824, USA; and 5Centre de Recherche en Cancérologie de Marseille, CNRS UMR7258, INSERM U1068, Ins;tut Paoli-CalmeSes, Aix-Marseille Université, Marseille, France; 6The Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9TJ, UK; 7Department of Biochemistry , University of Cambridge, Cambridge CB2 1GA, UK; 8current address, Department of Biotechnology, Dalian Medical University, Dalian 116044, China.*Correspondence: kmeek@msu.edu 25 ABSTRACT DNA double strand breaks (DSBs) are induced by external genotoxic agents (ionizing radiation or genotoxins) or by internal processes (recombination intermediates in lymphocytes or by replication errors). The DNA ends induced by these genotoxic processes are often not ligatable, requiring potentially mutagenic end-processing to render ends compatible for ligation by non- homologous end-joining (NHEJ). Using single molecule approaches, Loparo and colleagues propose that NHEJ fidelity can be maintained by restricting end-processing to a ligation competent short-range NHEJ complex that “maximizes the fidelity of DNA repair”. These in vitro studies show that although this short-range NHEJ complex requires DNA ligase IV (Lig4), its catalytic activity is dispensable. Here using cellular models, we show that inactive Lig4 robustly promotes DNA repair in living cells. Compared to repair products from wild-type cells, those isolated from cells with inactive Lig4 show a somewhat increased fraction that utilize micro- homology (MH) at the joining site consistent with alternative end-joining (a-EJ). But unlike a-EJ in the absence of NHEJ, a large percentage of joints isolated from cells with inactive Lig4 occur with no MH -- thus, clearly distinct from a-EJ. Finally, biochemical assays demonstrate that the inactive Lig4 complex promotes the activity of DNA ligase III (Lig3). 26 INTRODUCTION A growing consensus is emerging that DNA double-strand break repair by the non- homologous end-joining [NHEJ] pathway proceeds through distinct steps (3-10). Loparo and colleagues have shown that a long-range synaptic complex that positions the DNA ends ~115 Angstroms (Å) apart is dependent on the catalytic subunit of the DNA dependent protein kinase [DNA-PKcs, DNA-PK] and its regulatory subunit the DNA end-binding factor Ku. Transition of these long-range complexes to short-range synaptic complexes requires the catalytic activity of DNA- PK and the NHEJ ligase complex including XRCC4, XLF, and DNA ligase IV [Lig4]. These in vitro studies establish that whereas the long-range complex effectively blocks DNA end-processing, the short-range complex facilitates both end-processing and ligation (6,11). These studies are in good agreement from cellular studies from Ramsden and colleagues who propose that the ligase complex helps to limit end-processing to promote more error-free repair (12). Of note, in these in vitro studies, the catalytic activity of Lig4 is not required to either promote formation of the short-range complex or to facilitate end-processing that occurs in the complex. This result is consistent with previous studies proposing various structural roles for the Lig4 complex including promotion of end-processing (13), promotion of end-synapsis (2,14,15), and notably a report from Chiruvella et al who show that in yeast, a catalytically inactive Lig4 complex increases chromosomal end-joining (16). Support for this emerging two-step model of NHEJ has been bolstered by recent cryo-EM studies that precisely delineate potential long-range and short- range NHEJ complexes (7-9). In these previous studies, although end-processing mediated by the X family polymerases poll and polm, as well as TDP1, and PNKP was clearly dependent on the short-range complex, end-processing by the Artemis nuclease was not (11). Artemis functions exclusively with DNA- PKcs; its role in facilitating opening of the hairpin DNA termini associated with VDJ recombination [the process that provides for the generation of a diverse repertoire of antibodies and T cell receptors] has been well-defined (17-19). However, Artemis also functions to repair a subset of DSBs with non-ligatable DNA ends (20). Work from our laboratory also strongly suggests that Artemis hairpin opening is not restricted to the short-range complex (21). Briefly, using episomal end-joining assays, we found that cells which lack the NHEJ ligase complex robustly open hairpin 27 termini. These data are consistent with experiments in developing mouse lymphocytes from NHEJ deficient mice. In these studies, opened hairpin coding joints are observed in mice with defects in the ligase complex (22), but not in mice with defects in either DNA-PK or Artemis (23,24); but, coding end-joining is dramatically impaired in mice with any of these defects in NHEJ (23). Finally, these results are completely consistent with recent structural studies that demonstrate that DNA-PK and Artemis function in a monomeric complex that does not require the Lig4 complex that is apparently requisite for other end-processing activities (25). It has been known for decades that NHEJ defective cell lines robustly repair DNA DSBs on episomes using the alternative non-homologous end-joining pathway (a-EJ)(26,27). However, our recent study demonstrates that cells lacking the XRCC4/Lig4 complex are similarly defective in joining the opened hairpins as are cells lacking either DNA-PKcs or Artemis where the hairpins remain sealed (21). This suggests that the DNA-PK/Artemis complex that facilitates hairpin opening also, via some undefined mechanism, shields the opened hairpin ends from a-EJ; this “undefined mechanism” would be consistent with the function of the long-range complex (blocking end-processing and ligation) proposed by Loparo and colleagues (11). Here we explore the potential basis for Lig4's non-catalytic role in NHEJ. To test whether the presence of the ligase complex (but not its ligase activity) is sufficient to promote end- processing in living cells, we generated (via Crispr strategies) both Lig4 deficient and enzymatically inactive Lig4 mutant cell strains. We find that whereas Lig4 deficient cells are similarly impaired in joining DSBs with hairpin termini as are other NHEJ defective cells, cells expressing inactive Lig4 are remarkably proficient in rejoining these DSBs. We conclude that the DNA-PK complex and perhaps the long-range synaptic complex protects DSBs from other DNA ligases and end-processing factors (except for Artemis) in living cells. Moreover, our cellular experiments demonstrate that a catalytically inactive Lig4 complex can efficiently promote end- joining in living cells, consistent with the model that NHEJ-mediated end-processing is limited to a short-range synaptic complex. It follows that either Lig3 or Lig1 must be capable of facilitating ligation of DNA ends processed by NHEJ's short-range complexes. Thus, we used in vitro assays and establish that the catalytically inactive Lig4 complex robustly promotes the activity of Lig3 but not Lig1. Finally, repaired DSBs recovered from cells expressing catalytically inactive Lig4 have 28 increased utilization of short sequence micro-homologies (MH) at the joining site, a well- established characteristic of a-EJ (28). RESULTS Figure 5: Cells expressing catalytically inactive DNA ligase IV or completely deficient in DNA ligase IV are markedly sensitive to the DSB-inducing agents calicheamicin and teniposide. (A) Western blot of Ligase IV in wild-type 293T cells (Lig4+/+), Lig4 haplo-insufficient cells (Lig4+/), or 293T cells deficient in Lig4 (Lig4-/-), deficient in DNA-PKcs (DNA-PKcs-/-) or with mutations in Lig4 [K273A/-, and K273A*/-]. (These are two independent clones possessing one copy of catalytically inactive Lig4 and a frameshift mutation on the second Lig4 allele; the K273A clone containing an additional mutation, I270V is labelled K273A*/-, for brevity.) (B) Sensitivity of the same panel of cell strains to calicheamicin (B) or teniposide (C) was assessed. Briefly, cells were plated in 24 well plates into complete medium with increasing doses of calicheamicin or teniposide. After 7 days, MTT was added, and cell survival assessed by colorimetry. Survivakl assays were performed four times in duplicate (B) or three times in duplicate (C). Cells expressing catalytically inactive DNA ligase IV or completely deficient in DNA ligase IV are markedly sensitive to DSB-inducing agents. To begin to address whether Lig4 has a non-catalytic function in living cells, a CRISPR strategy was devised using a single gRNA that targets the codon AAG, encoding K273 [the well-conserved catalytic site (16,29,30) in Lig4 and a single stranded oligonucleotide to direct an HDR mediated K273A mutation at this site (as well as the introduction of a novel HhaI site). From these transfections in 293T cells, single clones were isolated, and DNA was isolated for PCR/HhaI digestion. Amplicon sequencing was performed on clones that included addition of the HhaI site in the initial PCR screen. Four clones were chosen for further analyses (Supplemental Table 1). Sanger sequencing from one clone (clone 17, that lacks a wild- 29 type allele by initial PCR screen) revealed two alleles with frameshift mutations, and we conclude that this clone is completely deficient in Lig4. Amplicon sequencing of two clones (that have a novel HhaI site ascertained by the initial PCR screen) reveal that one (clone 144) has the targeted K273A mutation on one allele, and a frameshift mutation on the second allele. The second clone (clone 195) contains the K273A mutation as well as an additional mutation, I270V on one allele, and the second allele includes a frameshift mutation (Supplemental Table 1). A heterozygous clone (clone 104) was isolated that has an inactivating frameshift mutation on one allele, but the second allele is wild-type. Cells with defects in the NHEJ pathway are markedly sensitive to numerous different DNA damaging agents that can induce DNA DSBs by various mechanisms. We chose two drugs, calicheamicin [generating DSBs with 3' overhangs with 3' phosphoglycolate (3'PG)] (31) that generates DSBs throughout the cell cycle (31) and teniposide a topoisomerase II (topII) inhibitor that generates two-ended DSBs primarily in S phase [with 5' overhangs with 5' hydroxyls] (32). Cellular calicheamicin and teniposide sensitivity was assessed for wild-type 293T as well as Lig4+/-, K273A/-, K273A+I270V/- (labelled K273A*/-, for brevity), and Lig4-/-, clones as well as a previously described DNA-PKcs deficient clone (33) using MTT staining as a measure of cellular viability (Figure 5B + C). As can be seen, cells completely deficient in Lig4 are remarkably sensitive to calicheamicin and teniposide; the two clones expressing only catalytically inactive Lig4 are also sensitive to both drugs, but substantially more resistant than cells that are completely deficient in Lig4. In contrast, at these doses, the Lig4+/- heterozygous clone is similarly resistant to both drugs as wild-type 293T cells. Finally, the previously described DNA-PKcs-/- 293T cells are also hypersensitive to calicheamicin and teniposide, but more resistant than Lig4-/- cells consistent with previous studies (23). These data suggest that catalytically inactive Lig4 promotes survival (albeit inefficient) to calicheamicin and teniposide. 30 Figure 6: Catalytically inactive DNA ligase IV promotes joining of DSBs in episomal substrates in 293T cells. (A) Fluorescent substrates (depicted in top panels indicate promoter with arrow) were utilized to detect TelN, ISce-1, or V(D)J coding and signal joints in 293T cells of the indicated genotypes. 293T clones with the indicated genotypes were co-transfected with TelN or I-SceI plasmid substrate, with and without enzyme (-/+) and analyzed for red/green fluorescence via flow cytometry (lower panels). (B) Lig4-/- 293T cells were tested for TelN and I-SceI joining by co-transfecting the TelN/I-SceI substrates and expression plasmids with or without co- transfection of expression plasmids encoding either WT or catalytically inactive human ligase IV as indicated. (C) 293T clones were co-transfected with wild-type (W) or hypermutant (M) RAG expression plasmids with either plasmid substrates to detect coding or signal end-joining as indicated by analysis of red/blue fluorescence via flow cytometry. Episomal V(D)J assays testing joining of coding (hairpin) and signal (blunt) ends. Cells were transfected with substrate and either: no Rag2 (-), WT rags (W), or mutant rag2 (M) and analyzed via flow cytometry. In A, B, and C, student's T test comparing joining rates between Lig4-/- and either Lig4+/+, Lig4+/-, or K273A were performed; ****P<0.0001; ***P<0.001; ns=not significant in two-tailed unpaired t test. 31 Catalytically inactive DNA ligase IV promotes joining of DSBs on episomal substrates in 293T cells. We have previously developed plasmid substrates with recognition sequences for DNA endonucleases that can produce a range of DNA end structures. The cutting sites flank the coding sequence for a red fluorescent protein (Crimson, RFP) driven by a CMV promoter and by co- transfecting substrate with appropriate enzymes, end-joining efficiency can be assessed by measuring the fraction of cells that express GFP or CFP after excision of the RFP cassette. The TelN protelomerase and I-SceI homing endonuclease produce hairpin DNA ends or 4 nucleotide overhangs respectively. To test whether 293T cells that either lack Lig4 entirely or retain a single copy of a catalytically inactive Lig4 gene can rejoin TelN or I-SceI-induced DSBs, a series of episomal end-joining assays were performed (Figure 6). Our previous work has shown that cells with defects in either the DNA-PK complex, Artemis, or the Lig4 complex are all significantly and similarly impaired in joining TelN-induced hairpin ends, even though the hairpinned ends are opened in cells deficient in the Lig4 complex (that retain DNA-PK and Artemis), but are obviously sealed in cells lacking DNA-PK or Artemis (21). Consistent with this previous study, cells deficient in Lig4 have a marked decrease in TelN joining (Figure 6A). In contrast, rejoining I-SceI DSBs is only ~50% reduced in cells deficient in Lig4, because restriction enzyme-induced DSBs are available to the a-EJ pathway [consistent with previous studies, (26,27,34)]. These data suggest that engagement of the Artemis/DNA-PK complex to resolve the closed hairpins must in some undefined way restrict the opened hairpins to the NHEJ pathway. Strikingly, in contrast to Lig4 deficient cells, the two independent clones expressing only catalytically inactive Lig4 rejoin significant levels of both I-SceI and TelN-induced DSBs (Figure 6A). These data suggest that the Lig4 complex (whether active or not) promotes end-joining by a-EJ presumably by relaxing the NHEJ restriction imposed by hairpin opening by DNA-PK/Artemis. To confirm that the observed differences in end-joining are the result of the targeted Lig4 mutations, we utilized the Lig4 deficient 293T cells and expression vectors encoding wild-type or catalytically inactive human Lig4 (Figure 6B) in a complementation experiment. As can be seen, both wild-type and catalytically inactive Lig4 (but not empty vector) substantially reverse both TelN and ISce-1 joining in 293T cells completely deficient in Lig4. 32 In cells that lack NHEJ, DSBs (including those induced by restriction enzymes, those introduced during class switch recombination, or by CRISPR or other gene editing strategies) can all be efficiently re-joined by the a-EJ pathway. In contrast, NHEJ-deficient cells do not join VDJ- associated DSBs because VDJ recombination intermediates are tightly restricted to the NHEJ pathway by mechanism(s) that are still incompletely understood. To corroborate the TelN joining experiments, we next assessed V(D)J coding and signal end-joining in the same panel of 293T cells. In these experiments we used both wild-type RAG expression vectors as well as a well- studied hyper-RAG2 mutant that substantially increases VDJ joining by de-stabilizing the RAG post-cleavage complex(s) that function to limit end-joining to the NHEJ pathway (35). As can be seen, 293T cells lacking Lig4 are effectively incapable of joining RAG-induced DSBs, either blunt- ended signal ends or hairpinned coding ends. In contrast, cells expressing catalytically inactive Lig4 join both coding and signal ends at levels only modestly reduced as compared to levels observed in wild-type 293T or Lig4 haplo-insufficient 293T cells. The hyper RAG2 mutant substantially increases the level of both coding and signal joining in wild-type cells, but joining is minimal in cells completely deficient in Lig4. In contrast, both signal and coding end-joining is robust, and only modestly reduced in cells expressing catalytically inactive Lig4 in experiments utilizing the hyper-RAG mutant (Figure 6C). It is interesting to note that cells expressing catalytically inactive Lig4 have a more severe deficit in signal end-joining than in coding end- joining. To our knowledge, this is unlike VDJ deficits in any other NHEJ mutant studied to date, and perhaps suggest that catalytically inactive Lig4 can promote joining of over-hanged DNA ends better than blunt DNA ends. In sum, we conclude that catalytically inactive Lig4 efficiently promotes rejoining of RAG-induced DSBs. These data reveal that once V(D)J intermediates are released to a complete Lig4 complex (potentially, the short-range NHEJ complex), the strict requirement for NHEJ-only dependent joining of RAG-induced DSBs has been fulfilled, and the RAG-induced DSBs can be joined by the a-EJ ligases just as well as non-RAG induced DSBs. 33 Figure 7: Catalytically inactive DNA ligase IV promotes joining of DSBs in episomal substrates in a variety of cell types. (A) Lig4-/- HCT116 cells were tested for TelN and I-SceI joining by co- transfecting the TelN/I-SceI substrates and expression plasmids with or without co-transfection of expression plasmids encoding either WT or catalytically inactive human ligase IV as indicated. (B) Lig4-/- U20S cells were tested for TelN and I-SceI joining by co-transfecting the TelN/I-SceI substrates and expression plasmids with or without co-transfection of expression plasmids encoding either WT or catalytically inactive mouse ligase IV as indicated. In A and B, student's T test comparing joining rates between vector and Lig4 or K273A were performed; ****P<0.0001; ***P<0.001; **P<0.01; ns=not significant in two-tailed unpaired t test. Catalytically inactive DNA ligase IV promotes joining of DSBs on episomal substrates in diverse cell types. To extend these studies, a CRISPR/Cas9 strategy was utilized to ablate Lig4 from the U2OS cell strain, and a Lig4 deficient HCT116 cell strain was obtained from Dr. Eric Hendrickson (36). As can be seen, in these cell types, both wild-type and catalytically inactive Lig4 (but not empty vector) substantially reverse both TelN and ISce-I end-joining (Figure 7), completely 34 analogous to the results observed in Lig4-/- 293T cells. These data substantiate our conclusion that the Lig4 complex facilitates end-joining of a variety of DSBs on episomal substrates. Figure 8: Catalytically inactive Ligase IV is only moderately deficient in CSR. (A) Western blot of murine Ligase IV expressed in Lig4-/- CH12 cells. (B) CSR efficiency (percentage of IgA-positive cells, assessed by flow cytometry) in mouse CH12 cells expressing WT ligase IV, catalytically inactive Ligase IV (dLig4), or no ligase IV. Error bars indicate SE of three independent experiments. (C) Sensitivity of the same panel of cell strains to calicheamicin was assessed. Briefly, cells were plated in 24 well plates into complete medium with increasing doses of calicheamicin. After 7 days, MTT was added, and cell survival assessed by colorimetry. In B, student's T test comparing joining rates between vector and Lig4, or vector K273S were performed; ****P<0.0001; in two- tailed unpaired t test. Survival assay was performed three times in duplicate. Catalytically inactive DNA ligase IV supports chromosomal rearrangement during class switch recombination. Previously we studied the role of Lig4 in the process of immunoglobulin class switch recombination (CSR) using a well-studied mouse B cell model, CH12 cells (37). Retroviral vectors encoding either wild-type or K273S murine Lig4 were prepared and used to transduce a previously described Lig4-/- Ch12 cell strain. When CH12 cells are stimulated with an anti-CD40 antibody, interleukin 4, and TGFβ1 (+CIT), CSR (from IgM to IgA) is robustly induced. This produces clear populations of IgM+/IgA-, IgM+/IgA+, and IgM-/IgA+ cells that can be analyzed by flow cytometry. Like VDJ recombination, CSR is a lymphocyte specific DNA recombination event that proceeds through a double-strand break intermediate; but unlike VDJ recombination, a-EJ can facilitate CSR (albeit at reduced levels) in the absence of most NHEJ factors. Our previous study demonstrated that either Lig1 or Lig3 could facilitate a-EJ in a Lig4 deficient murine B cell line (CH12 cells) that were induced to undergo class switch recombination. 35 We next assessed CSR in the CH12 Lig4-/- cells expressing wild-type Lig4, K273S, or vector only from lentivirus expression vectors. Consistent with our previous study, only ~10% of Lig4 deficient CH12 cells expressing vector alone undergo CSR compared to ~60% of cells expressing wild-type murine Lig4. Cells expressing K273S Lig4 display an intermediate level of CSR (Figure 8). We conclude that catalytically inactive Lig4 promotes chromosomal end-joining of cell- programmed DSBs. We also assessed cellular resistance to calicheamicin in this panel of CH12 cells. As can be seen, whereas cells expressing wild-type Lig4 are substantially more resistant to calicheamicin than cells expressing no Lig4, cells expressing catalytically inactive Lig4 are similarly sensitive to calicheamicin as are cells lacking Lig4. We conclude that catalytically inactive Lig4 cannot restore cellular resistance to DNA damaging agents in all cell types. Figure 9: CatalyZcally inacZve Lig4/XRCC4/XLF promotes Lig3-mediated joining of both cohesive and blunt DNA ends in vitro. The capacity for catalygcally inacgve Lig4/XRCC4 to promote ligagon by Lig3 (A), or Lig1 (B) was assessed by incubagng a blunt DNA ligagon substrate (linearized pUC19) with purified recombinant proteins as indicated at the following concentragons: Ku, 250nM; XLF, 200nM; LIG4/XRCC4, 500nM; LIG3/XRCC1, 40nM; and LIG1,180nM. 10ul reacgons were incubated for 30 minutes at room temperature and analyzed by gel electrophoresis and quangfied with Image J. Bar graph represents three independent experiments and error bars represent SD. 36 Catalytically inactive Lig4/XRCC4/XLF promotes Lig3-mediated joining of both cohesive and blunt DNA ends in vitro. The robust joining facilitated by catalytically inactive Lig4 in cellular assays suggests that the NHEJ short-range complex might be capable of facilitating ligation of DNA ends by either Lig3 or Lig1. To directly test this possibility, Lig1 and Lig3/XRCC1 were expressed and purified from E. coli (Sup. Figure 1A) and tested for activity (Sup. Figure 1B). Then purified Lig1 or Lig3/XRCC1 were tested in ligase assays that included the components of the NHEJ short range complex (Ku, X4/Lig4, and XLF) using catalytically inactive Lig4, on substrates with either blunt or cohesive DNA ends. In these assays, higher levels Lig4/XRCC4 were used as compared to either Lig1 (2.8 molar excess) or Lig3/XRCC1 (12.5 molar excess). As can be seen (Figure 9A), using a blunt ligation substrate, no ligation products are observed with just the NHEJ components, and no activity is observed with only Lig3/XRCC1. In contrast, when Lig3/XRCC1 is added to reactions containing catalytically inactive Lig4 (with or without Ku), robust ligation is observed. This activity is completely dependent on both Lig4/XRCC4 and XLF, but Ku is dispensable. In parallel assays, Lig1 activity is not significantly enhanced by catalytically inactive Lig4 complex (Figure 9B) under any of the conditions tested. This assay was repeated using DNA with cohesive ends (Sup. Figure2); as can be seen, ligation of compatible ends is robust in the presence of catalytically inactive Lig4 complex and Lig3/XRCC1, but not Lig1, suggesting that catalytically inactive Lig4 complex can stimulate Lig3/XRCC1-mediated joining of cohesive ends. However, (to our surprise) we consistently observed very low levels of ligation products in reactions with XLF and XRCC4/Lig4 (catalytically inactive, K273A) without Lig3/XRCC1 or Lig1. The activity represents <5% the activity observed in assays with wild-type Lig4 complexes (compare Sup. Figure 1C and 2A). This residual activity could not be attributed to a contaminating ligase since the proteins are prepared in E. coli that has only NAD dependent ligase. Moreover, adenylation assays demonstrate minimal adenylation of the K273A mutant Lig4 (Sup. Figure 3). From the recent cryo-EM study of the short-range complex (7), we observed that 4 additional lysine residues are very close to the 5’ phosphate of the DNA end; whereas K273 is ~7Å away, lysines 449, 451, 352, and 345 are between 8Å and 9.5Å away from the 5’ phosphate. We considered that one of these four lysines might serve as a back-up adenylation site explaining the minimal ligase activity of the K273A mutant. These four residues 37 were substituted with arginine in the K273A mutant construct. As can be seen, this 5X lysine mutant has no residual activity in ligase assays but retains the capacity to stimulate Lig3 activity towards both blunt and cohesive ends in vitro (Supplemental Figure 4A) to a similar extent as the K273A mutant. Moreover, in cellular assays, the 5X lysine mutant joining activity that is indistinguishable from that of the K273A mutant (Supplemental Figure 4B). From these data, we conclude that the catalytically inactive Lig4/XRCC4/XLF complex promotes end-joining of Lig3 in vitro, potentially explaining the robust joining activity observed in living cells expressing the catalytically inactive complex. Figure 10: DSBs repaired in cells expressing K273A Lig4 have characteristics of a-EJ. (A) 293T clones of the indicated genotypes were transfected with Fill-in joining substrate either uncleaved, or cleaved with Eco53KI and PspOMI (to generate blunt and 5' overhangs), or Eco53K1 and Apa1(to generate blunt and 3' overhangs). Cells were assessed for RFP and GFP expression 72- 38 Figure 10 (cont’d) hours later. Student's T test comparing joining rates between Lig4-/- and either Lig4+/+, Lig4+/-, or K273A*/- were performed; ****P<0.0001; in two-tailed unpaired t test. (B) 293T clones of the indicated genotypes were co-transfected with the alt-VDJ substrate, a ds-RED expression plasmid, and hypermutant RAG expression plasmids. Restoration of the GFP reading frame requires joining via a 9bp region of MH that occurs 10bp from the termini of both coding ends. Cells were assessed for RFP and GFP expression 72 hours after transfection. Student's T test comparing joining rates between Lig4-/- and either Lig4+/+, Lig4+/-, or K273A*/- as well as betwee Lig4+/+ and either Lig4+/- or K273A*/- were performed; ****P<0.0001; ***P<0.001; **P<0.01; in two-tailed unpaired t test. Cells expressing K273A Lig4 cannot perform fill-in end-processing, but robustly promote a-EJ in episomal assays. Clearly catalytically inactive Lig4 enhances end-joining of episomal substrates and in some cell types, cellular survival after exposure to agents that induce DSBs. To gain knowledge as to how end-joining is enhanced by the catalytically inactive Lig4 complex, we used assays that clarify structural characteristics of repaired DSBs. More specifically, we focused on determining whether catalytically inactive Lig4 promotes end-joining that is similar to authentic NHEJ [i.e. generating joints ranging from perfect end-joining with no or minimal base pair loss, or joining at sites of MH (1-3bp)], or alternatively, if catalytically inactive Lig4 promotes joining that is more similar to a-EJ [characterized by increased loss of terminal nucleotides, and a strong dependence on the presence of MH (often longer than 3bp)]. Although NHEJ is generally characterized as an error-prone repair mechanism, numerous studies have documented the fidelity of NHEJ when joining most DSBs (11,12). For example, perfect joining of compatible ends are highly favored and incompatible ends are rejoined to minimize nucleotide loss (11,12). To assess the capacity of catalytically inactive Lig4 to promote joining that is similar to that of NHEJ, a joining substrate that measures fidelity of joining was generated (Figure 10A). Briefly, the substrate plasmid is restricted with two enzymes to generate a blunt end on one side and an over-hanged end (either 5' or 3') on the other side as illustrated. To restore the crimson open reading frame, the ends must be aligned so that the missing bases across from the overhangs are filled in. In both cellular and in vitro models of NHEJ, perfect fill-in of DNA ends like these is efficiently mediated by NHEJ (11,12). If uncut plasmid is transfected, crimson is not expressed because of the disruption in the open reading frame, but GFP is expressed by use of its own ATG. When restricted plasmids are transfected, if any plasmid 39 rejoining occurs, GFP is expressed. With this assay, "Perfect rejoining" represents the percent GFP positive cells that also express crimson, and in these assays, GFP expression is robust in all cell types. As would be expected, cells completely lacking Lig4-/- are completely unable to perfectly rejoin the transfected substrate promoting crimson expression, whereas in wild-type 293T cells more than 30% of the cells that rejoin the plasmid, rejoin the plasmid perfectly and crimson is robustly expressed (Figure 10A). In contrast to other joining assays (Figure 6), cells expressing only catalytically inactive Lig4 are similarly deficient in perfect rejoining as are cells completely lacking Lig4. We conclude that catalytically inactive Lig4 cannot promote perfect rejoining of the fill-in episomal substrate. It follows that the catalytically inactive complex either does not efficiently support fill-in end-processing, or that the cellular ligase facilitating end- joining (Lig3 or Lig1) does not efficiently join the filled-in ends. To assess whether a-EJ-like events facilitate repair in cells expressing catalytically inactive Lig4, we utilized an assay developed by Roth and colleagues (38). It is well-appreciated that short sequence homologies at opened coding-end termini can facilitate coding end-joining (39); these short sequence homologies are generally small (1-3 base pair) and occur close to the DNA terminus. We utilized their a-EJ assay (Figure 10B, termed alt-VDJ, using mutant RAG expression constructs that destabilize the RAG post-cleavage complex(s) to promote more alt-VDJ) to determine whether cells expressing catalytically inactive Lig4 depend on MH for joining. This assay requires nucleotide deletions of 10bp from each coding end, and then use of 9 base pair of short sequence homology to restore the GFP open-reading frame. Whereas minimal alt-VDJ is observed in wild-type 293T cells or Lig4 haplo-insufficient cells where end-processing generally precludes loss of the required 10bp from each coding-end, alt-VDJ is readily detected in Lig4-/- 293T cells (Figure 10B). Strikingly, alt-VDJ joining is robust in cells expressing catalytically inactive Lig4; these data suggest that K273A mutant Lig4 promotes end-joining that is similar to a-EJ. 40 Figure 11: Rejoined episomal VDJ coding and signal joints in cells expressing K273A Lig4 have characteristics of a-EJ mediated joining. (A) Coding (left) and signal joints (middle) were PCR amplified from substrates isolated from cells of the indicated genotypes 72 hours after transfection. (*) indicates a PCR artifact. Half of the PCR reaction for signal joint amplification was digested with ApaLI prior to electrophoresis (right). (B) Isolated coding joints from the indicated cell strains were subjected to amplicon sequencing. Histograms depicting numbers of nucleotides deleted or extent of microhomology at site of joining. 41 Rejoined episomal DSBs in cells expressing K273A Lig4 have characteristics of a-EJ mediated joining. To further characterize DSB joining events in cells expressing catalytically inactive Lig4, we characterized VDJ joints isolated from cells expressing the inactive complex. In cells with intact NHEJ, VDJ coding end-joining generates a diverse array of rejoined opened hairpin coding ends. In contrast, rejoining of the blunt signal ends is usually a precise, perfect head-to-head joining of the two heptamers. This recapitulates VDJ joining characteristics in developing lymphocytes. It has been shown that the rare VDJ joints generated in NHEJ deficient cells or animals have characteristics of a-EJ, such that coding joints have increased levels of nucleotide loss and the joints often occur at regions of MH; whereas signal ends are not perfectly rejoined. As shown above (Figure 6C), in cells completely deficient in Lig4, the joining rate of both coding and signal ends is severely reduced. Coding and signal joints from episomal assays were PCR amplified and analyzed by gel electrophoresis. As can be seen (Figure 11A, left), coding joints amplified from wild-type or Lig4 haplo-insufficient 293T cells, generate a diverse “smear” of coding joints, whereas coding joints amplified from 293T cells that express only catalytically inactive Lig4 are homogeneous, and slightly smaller than those recovered from wild-type cells. As expected, signal joints from wild-type or Lig4 haplo-insufficient 293T cells are uniform, but signal ends isolated from cells that express catalytically inactive Lig4 are diverse, generating a smear of joints (Figure 11A middle). Rejoining of signal ends (without nucleotide loss or gain) generates a novel ApaLI site; so fidelity of signal end-joining can be ascertained by restricting signal joints with this enzyme. Signal joints amplified from assays in wild-type cells are uniform and largely susceptible to ApaLI cleavage consistent with perfect rejoining of signal joints in NHEJ- proficient cells (40). In contrast, signal joints amplified from cells that lack Lig4 or express only catalytically inactive Lig4 are completely resistant to ApaLI cleavage indicating nucleotide loss or addition from the signal ends prior to rejoining, suggesting catalytic inactive Lig4 complex is not proficient at promoting perfect blunt end-joining (Figure 11A, right). PCR amplified coding joints were submitted for amplicon sequencing; as can be seen (Figure 11B), joints isolated from wild-type or Lig4 haplo-insufficient cells are virtually indistinguishable, and ~50% do not have sequence MH at the site of joining. In these assays, there is a bias for joining at particular sites of microhomology resulting over-representation of 42 sequences with 3bp and 5bp deletions; these over-represented joints can be appreciated in all three samples. In wild-type cells, the predominate 3bp and 5bp deletion products account for 5.99% and 12.12% of all sequences, in L4 haplo-insufficient cells, 9.28% and 14.66%, but in K273A mutant cells these two particular joints account for 27.4% and 18.37% of all joints. These data support the conclusion that joints facilitated by catalytically inactive Lig4 have characteristics of a-EJ. Figure 12: Chromosomal DSBs repaired in cells expressing K273A Lig4 have characteristics of a- EJ. (A) Diagram of region on chromosome nine targeted by two different gRNAs and position of primers utilized to detect chromosomal deletions induced by Cas9 and indicated gRNAs. (B) Summary of amplicon analyses of PCR amplified deletional joints from indicated cell strains. (C) Histograms depicting numbers of nucleotides deleted, inserted, or extent of microhomology at site of joining. 43 Chromosomal end-joining in cells expressing K273A Lig4 has characteristics of a-EJ. To extend these findings to chromosomal end-joining, we exploited CRISPR/Cas9 targeting of the FANCG gene on chromosome 9, which we found to be remarkably efficient in 293T cells (41). Although this strategy does not allow quantification of joining, the quality of joining can be assessed by sequencing. Briefly, cells of the indicated genotypes were transfected with two gRNA/cas9/puro plasmids that target sequences ~300bp apart; after 48 hours, cells were subjected to puromycin selection. After 72 hours, genomic DNA was isolated and PCR was utilized to assess deletional rejoining of the two DSBs. Isolated PCR fragments were submitted for amplicon sequencing. In wild-type cells, ~92% of recovered joints are perfect (of the blunt-ended Cas9 cleavage sites), whereas in K273A mutant cells, only ~80% of the joints are perfect. In wild-type cells, the average nucleotide loss/joint is 0.45bp, and ~4% of joints occur at sites of MH. In contrast, in K273A mutant cells, the average loss/joint was 2.9bp, and ~19% occur at sites with MH. The range of nucleotide loss, nucleotide insertion, and utilization of short sequence homologies for joints from wild-type or K273A mutant cells is shown in Figure 12C. We conclude that joints from K273A mutant cells are consistent with joining via a-EJ. DISCUSSION In other mutational studies that block enzymatic function of NHEJ factors (ie Artemis or DNA-PKcs), inactivation results in a cellular phenotype that is similar to (42-44) or in certain cell types worse than complete loss (33,45). Thus, it appears that the Lig4 complex is unique in that in its inactive form, Lig4 can promote the function of other repair factors. There have been several previous studies proposing various structural roles for the Lig4 complex. In 2007, Chu and colleagues reported that the NHEJ ligase complex promotes end- processing activities in vitro (13). This is consistent with more recent reports, where in an in vitro model of NHEJ, Stinson et al. demonstrated that Lig4 (either active or inactive) was necessary for formation of a short-range non-homologous end-joining complex that closely juxtaposed two DNA ends, promoting both end-processing and ligation (11). Besides the studies from Loparo and colleagues suggesting a two-stage model of NHEJ (6,11), several other reports support a role in DNA end synapsis for the Lig4 complex (2,14,15). Reid et al demonstrated that the Lig4 complex can promote end-synapsis in vitro (using purified 44 proteins in a FRET assay), and the ability to synapse the ends was strongly impacted by DNA end structure, especially by the presence of the 5’ phosphate (2). Of note, a K273A mutant Lig4 complex was defective in synapsing DNA ends in their assays using purified proteins (2), whereas similar FRET assays from Graham et al showed that K273A using Xenopus extracts, fully support end synapsis (6). Conlin et al, extended the studies of end synapsis to show that a unique pocket in Lig4 allows synapsis of DNA ends with mis-matched termini (15). There are also cellular studies that support a role for the Lig4 complex in DNA end synapsis. Cottarel et al. demonstrated that cells deficient in Lig4 are defective in damage-induced autophosphorylation of DNA-PKcs at serine 2056 (S2056) (14). We first showed that S2056 autophosphorylation could occur in trans, and that S2056 phosphorylation occurs almost exclusively by autophosphorylation (46). Recent structural studies strongly support the conclusion that S2056 phosphorylation occurs in trans (25). Cottarel et al. demonstrated that catalytically inactive Lig4 could restore the deficit in S2056 phosphorylation in Lig4 deficient cells as well as wild-type Lig4. From these studies they proposed that the catalytically inactive Lig4 complex facilitates synapsis, promoting trans- autophosphorylation of DNA-PKcs at S2056 (14). We extended this finding by demonstrating that XLF also participates in the capacity of the Lig4 complex to promote S2056 phosphorylation in living cells, and that only XLF that was proficient in interacting with XRCC4 (to generate DNA- bridging filaments) could promote S2056 phosphorylation (47). Finally, Chiruvella et al reported that a catalytically inactive Lig4 complex increases not only synapsis, but chromosomal end- joining in yeast; this impact on end-joining was absolutely dependent on XLF (16). This result is entirely consistent with cellular assays presented here, and with the in vitro assays demonstrating that catalytically inactive Lig4/XRCC4 only stimulates Lig3/XRCC1 joining in the presence of XLF (Figure 9). Our working model is that Lig4 itself is important in promoting synapsis and that formation of a short-range synaptic complex (even one that lacks Lig4 activity): 1) facilitates joining of many DSBs, 2) promotes cellular resistance to drugs that induce DSBs in some cell types, and 3) fulfills the RAG-induced restriction step of VDJ recombination so that even RAG-induced DSBs (both coding and signal ends) can be joined by non-NHEJ DNA ligases. 45 In sum, data presented here extend these previous studies demonstrating a non-catalytic role for Lig4 that robustly stimulates both end-joining and cellular resistance to DNA damage. Joining that is facilitated by the inactive Lig4 complex shares characteristics with a-EJ (increased nucleotide loss, and increased use of microhomologies). This observation presents an unanticipated possibility, that the inactive Lig4 complex (likely consistent with short-range complexes proposed recently) may facilitate a-EJ mediated by Lig3. These studies present an important unanswered question: Do complexes with catalytically active Lig4 utilize Lig3 to facilitate joining in normal cells? Work is ongoing to address this possibility. MATERIALS AND METHODS Cell culture, genome editing, and survival assays. 293T, U2OS and HCT116 cells were cultured in Dulbecco’s Modified Eagle Medium (Life Technologies) supplemented with 10% fetal bovine serum (Atlanta Biologicals, GA), 2 mM L-glutamine, 0.1 mM non-essential amino acids, 1 mM sodium pyruvate, 100 U/ml penicillin, 100 µg/ml streptomycin (Life Technologies) and 10 µg/ml ciprofloxacin. CH12F3 cells were cultured in RPMI 1640 medium supplemented with 10% (vol/vol) FBS and 50 μM β-mercaptoethanol. Cas9-targeted gene disruption was performed using methods similar to those reported by Mali et al. (48). Briefly, duplex oligonucleotides (Integrated DNA Technologies) generating a gRNA specific for a PAM site targeting the K273 codon were cloned into pCas2A-puro. Cells were transfected with 2ug plasmid and a 120 nucleotide oligonucleotide encoding a K273A mutation as well as a silent mutation generating a novel restriction site (HhaI) (see Supplemental Table I). 48 hours after transfection, cells were replated at cloning densities in media containing puromycin (1ug/ml). Puromycin was removed after 72 hours. Isolated clones were selected, and DNA isolated with DNAzol (Sigma) according to the manufacturer’s protocol. Restriction digestion of PCR products was utilized to detect the K273A mutation. Western blotting was used to confirm expression and genotypes were confirmed by Sanger sequencing or Amplicon sequencing (Genewiz) as depicted in Supplemental Table 1. MTT staining was performed to assess cell viability for both 293T cells and CH12. 30,000 to 50,000 cells were plated in each well of a 24-well plate, containing medium with varying concentrations of zeocin. After 5 to 7 days of calicheamicin treatment, cells were treated with 1 46 mg/ml MTT (Sigma) solution for 1 hr. Medium containing MTT was then removed and formazan crystals thus produced were solubilized in acidic isopropanol. Absorbance was read at 570 nm to determine relative survival. Episomal end-joining assays. The fluorescent VDJ coding, signal, and alt-VDJ substrates have been described (21,38,49). The Fill-in substrate was generated by synthesizing (Integrated DNA Technologies) a fragment encoding crimson with the addition of restriction endonuclease sites Eco53KI, PspOMI, and Apa1 that disrupt the Crimson open-reading frame. Cleavage with these restriction enzymes generate blunt and over-hanged ends as described in Figure 11D. The Crimson fragment included NheI and BamHI which were used to subclone this fragment between the promoter and GFP open reading frame in the substrate plasmids. Briefly, extrachromosomal fluorescent joining assays were performed on cells plated at 20-40% confluency into 24-well plates in complete medium. Cells were transfected with 0.125 µg substrate, and either 0.25 I- Sce1, TelN, or RAG 1+2 expression plasmids per well using polyethylenimine (PEI, 1 ug/mL, Polysciences) at 2 µL/1 µg DNA. Cells were harvested 72 hours after transfection and analyzed for GFP and RFP expression by flow cytometry. The percentage of recombination was calculated as the percentage of live cells expressing GFP divided by the percentage expressing RFP. Data presented represents at least three independent experiments, which each includes triplicate transfections. In figure 11, the coding joint substrate was modified so that the coding flanks included only A or T sequences; in addition, the 23RSS was inverted to provide for inversional joining which facilitates PCR amplification of coding joints. Transfected plasmids were isolated by alkaline lysates 72 hours after transfection. Coding joints from each transfection were PCR amplified and analyzed by electrophoresis and amplicon sequencing by Genewiz. Class switch recombination assays. CSR assays were performed as described previously (37). Briefly, cells were seeded in the presence of 1 μg/mL anti-CD40 antibody (16-0402-86; bioscience), 5 ng/mL of IL-4 (404-ML; R&D Systems), and 0.5 ng/mL TGF-β1 (R&D Systems 240-B) and grown for 72 h. Cells were stained with a FITC-conjugated anti-mouse IgA antibody (BD Bio- sciences 559354) and analyzed on a LSR II flow cytometer (BD Biosciences). CSR efficiency is determined as the percentage of IgA-positive cells. 47 Amplicon sequencing of Crispr/Cas9-induced chromosomal DSBs. We have used a Crispr/Cas9 strategy to sequence DSBs from within the FancG locus previously (41). To induce deletional DSBs, two gRNA/Cas9/puro plasmids were transfected into 293T cells with the indicated genotypes. After 48 hours, cells were place in puromycin selection media. Cells were harvested and DNA prepared 72 hr later and PCR performed to detect chromosomal deletions. PCR fragments were isolated and subjected to Amplicon sequencing provided by Genewiz. Mammalian expression vectors and recombinant protein expression constructs and purification. Expression plasmids and purification procedures for XRCC4-WT, XLF-WT, Lig4-WT, and Lig4-K273A have been described (41).The LIG4/XRCC4 complex was produced as described (14). All proteins batches were dialyzed against 150 mM KCl, 20 HEPES pH8, 1 mM EDTA, 2 mM DTT and 10% (v/v) glycerol, flash frozen in liquid nitrogen and stored at -80 ̊C. T4 DNA ligase was obtained from New England Biolabs. LIG4 fragments 1-620 (WT or K273A), DBD, NTase, and OBD were prepared as described (50). Mammalian expression constructs for wild-type Lig4 and Lig4- K273A were generated by subcloning from plasmids described above. The 5XLys mutant was prepared by subcloning a geneblock (IDT) encoding K273A, K449R, K451R, K352R, and K345R via PflMI/BlpI subcloning. Ligation Assay. Ligation assays were performed in a volume of 10 μL containing 50 ng of linearized pUC19 plasmid DNA (with XbaI for cohesive ends and SmaI for blunt ends), 1 mM ATP, 1 mM DTT, 20 mM Tris-HCl pH8.0, 2 mM MgCl2, and 60 mM KCl, 1% (v/v) glycerol with addition of proteins at the indicated concentrations. Reactions were incubated at room temperature for 30 min before addition of Proteinase K at 1.4 mg/mL final concentration and 1X of a loading buffer containing 0.01% SDS (NEB #B7024S) and incubated for 10 min at 55°C. Samples were next resolved by 0.8% agarose gel electrophoresis in Tris-Borate-EDTA buffer. Gels were stained in Tris-Borate-EDTA buffer supplemented with 0.5 μg/mL ethidium bromide and destained in deionized water. Images where captured under UV light using a Bio-Rad chemidoc and quantitatively analyzed with Image J. Oligonucleotides and antibodies. Oligonucleotides used in this study are as follows; only one strand of the oligonucleotides used for targeting PAM sites are presented, and are without BMHI overhangs. 48 For 293T Lig4 CRISPR experiments: gRNA Lig4: CATACGTTCACCATCTAGCT Lig4 K273A HDR: CTATTGCAGATATTGAGCACATTGAGAAGGATATGAAACATCAGAGTTTCTACATAGAAACAGCGCTAG ATGGTGAACGTATGCAAATGCACAAAGATGGAGATGTATATAAATACTTCTCTCGAAATGG For U2OS CRISPR experiments: gRNA Lig4: GTTCAGCACTTGAGCAAAAG. The U20S clone used in the experiments had +1 frameshift mutations on both alleles. For 293T polQ CRISPR experiments: gRNA1 polQ:TGAAGCGGGTTTTGGAAATG gRNA2 polQ:TCTGATCAATCGCCTCATAG For 293T FANCG CRISPR experiments: gRNA1 FancG:GGGCCAGGCCTGGGTTCAAC gRNA2 FancG:GACTTAAGAGAAAGGGACTG 5’ FancG PCR: CCCAAGATGTCCCGGCTGTGGG 3’FancG PCR:CCATGGGCCTCTCTGTCCTTGCAC Oligonucleotides for substrate PCR: 5’ coding joint PCR: CGGTGGGAGGTCTATATAAGCA 3’ coding joint PCR:CTACACCGTGGTGGAGCAGTA 5’ signal joint PCR:ACCTTGAAGCGCATGAAGGGC 3’ signal joint PCR:TCCATGCGGTACTTCATGGTC Antibodies utilized in this study include: anti-DNA Lig 4 (Abcam, 26039), anti-Ku80 (Invitrogen, 111), anti-DNA-PKcs (generous gift Tim Carter). Data availability: The authors confirm that the data supporgng the findings of this study are available within the argcle and its supplementary materials; the amplicon sequencing files from this study are available from the corresponding author. Funding: USDA Nagonal Insgtute of Food and Agriculture [1019208]; Public Health Service [AI048758, AI147634 to K.M.] and [AI 38345, AI 39039 to KY]; French Nagonal Research Agency 49 (ANR-17-CE12-0020-01, ANR-18-CE29-0003-04) and French Nagonal League against cancer (équipe labellisée) to M.M. Acknowledgements: We are pargcularly indebted to both Dale Ramsden and Adam Luthman for their generous and invaluable assistance in analyzing amplicon sequencing projects for differences in juncgonal diversity and uglizagon of short sequence homologies. ContribuZon by the author (CMB dissertaZon requirement): This work contains work from mulgple authors. As first author, Noah Goff performed the bulk of the data collecgon, analysis, and contributed to the conceptualizagon/wrigng/edigng of this manuscript—with the excepgon of Figure 9 and supplemental figures 1-3. Note the supplemental figures are available in the online version of this manuscript at Nucleic Acids Research. 50 1. 2. 3. 4. 5. 6. 7. 8. 9. REFERENCES Reid, D.A., Keegan, S., Leo-Macias, A., Watanabe, G., Strande, N.T., Chang, H.H., Oksuz, B.A., Fenyo, D., Lieber, M.R., Ramsden, D.A. et al. (2015) Organizagon and dynamics of the nonhomologous end-joining machinery during DNA double-strand break repair. Proceedings of the Na9onal Academy of Sciences of the United States of America, 112, E2575-2584. Reid, D.A., Conlin, M.P., Yin, Y., Chang, H.H., Watanabe, G., Lieber, M.R., Ramsden, D.A. and Rothenberg, E. (2017) Bridging of double-stranded breaks by the nonhomologous end-joining ligagon complex is modulated by DNA end chemistry. Nucleic acids research, 45, 1872-1878. Zhao, B., Watanabe, G., Morten, M.J., Reid, D.A., Rothenberg, E. and Lieber, M.R. (2019) The essengal elements for the noncovalent associagon of two DNA ends during NHEJ synapsis. Nature communica9ons, 10, 3588. Jackson, S.P. (2002) Sensing and repairing DNA double-strand breaks. Carcinogenesis, 23, 687-696. Yoo, S. and Dynan, W.S. (1999) Geometry of a complex formed by double strand break repair proteins at a single DNA end: recruitment of DNA-PKcs induces inward translocagon of Ku protein. Nucleic acids research, 27, 4679-4686. Graham, T.G., Walter, J.C. and Loparo, J.J. (2016) Two-Stage Synapsis of DNA Ends during Non-homologous End Joining. Molecular cell, 61, 850-858. Chen, S., Lee, L., Naila, T., Fishbain, S., Wang, A., Tomkinson, A.E., Lees-Miller, S.P. and He, Y. (2021) Structural basis of long-range to short-range synapgc transigon in NHEJ. Nature, 593, 294-298. Chaplin, A.K., Hardwick, S.W., Stavridi, A.K., Buehl, C.J., Goff, N.J., Ropars, V., Liang, S., De Oliveira, T.M., Chirgadze, D.Y., Meek, K. et al. (2021) Cryo-EM of NHEJ supercomplexes provides insights into DNA repair. Molecular cell, 81, 3400-3409 e3403. Chaplin, A.K., Hardwick, S.W., Liang, S., Kefala Stavridi, A., Hnizda, A., Cooper, L.R., De Oliveira, T.M., Chirgadze, D.Y. and Blundell, T.L. (2021) Dimers of DNA-PK create a stage for DNA double-strand break repair. Nature structural & molecular biology, 28, 13-19. 10. Wang, J.L., Duboc, C., Wu, Q., Ochi, T., Liang, S., Tsutakawa, S.E., Lees-Miller, S.P., Nadal, M., Tainer, J.A., Blundell, T.L. et al. (2018) Dissecgon of DNA double-strand-break repair using novel single-molecule forceps. Nat Struct Mol Biol, 25, 482-487. 11. Sgnson, B.M., Moreno, A.T., Walter, J.C. and Loparo, J.J. (2020) A Mechanism to Minimize Errors during Non-homologous End Joining. Molecular cell, 77, 1080-1091 e1088. 12. Waters, C.A., Strande, N.T., Pryor, J.M., Strom, C.N., Mieczkowski, P., Burkhalter, M.D., Oh, S., Qaqish, B.F., Moore, D.T., Hendrickson, E.A. et al. (2014) The fidelity of the ligagon step 51 13. 14. 15. 16. 17. determines how ends are resolved during nonhomologous end joining. Nature communica9ons, 5, 4286. Budman, J., Kim, S.A. and Chu, G. (2007) Processing of DNA for nonhomologous end- joining is controlled by kinase acgvity and XRCC4/ligase IV. The Journal of biological chemistry, 282, 11950-11959. Cojarel, J., Frit, P., Bombarde, O., Salles, B., Negrel, A., Bernard, S., Jeggo, P.A., Lieber, M.R., Modesg, M. and Calsou, P. (2013) A noncatalygc funcgon of the ligagon complex during nonhomologous end joining. The Journal of cell biology, 200, 173-186. Conlin, M.P., Reid, D.A., Small, G.W., Chang, H.H., Watanabe, G., Lieber, M.R., Ramsden, D.A. and Rothenberg, E. (2017) DNA Ligase IV Guides End-Processing Choice during Nonhomologous End Joining. Cell reports, 20, 2810-2819. Chiruvella, K.K., Liang, Z., Birkeland, S.R., Basrur, V. and Wilson, T.E. (2013) Saccharomyces cerevisiae DNA ligase IV supports imprecise end joining independently of its catalygc acgvity. PLoS gene9cs, 9, e1003599. Lu, H., Shimazaki, N., Raval, P., Gu, J., Watanabe, G., Schwarz, K., Swanson, P.C. and Lieber, M.R. (2008) A biochemically defined system for coding joint formagon in V(D)J recombinagon. Molecular cell, 31, 485-497. 18. Ma, Y., Pannicke, U., Schwarz, K. and Lieber, M.R. (2002) Hairpin opening and overhang processing by an Artemis/DNA-dependent protein kinase complex in nonhomologous end joining and V(D)J recombinagon. Cell, 108, 781-794. 19. 20. Goodarzi, A.A., Yu, Y., Riballo, E., Douglas, P., Walker, S.A., Ye, R., Harer, C., Marche~, C., Morrice, N., Jeggo, P.A. et al. (2006) DNA-PK autophosphorylagon facilitates Artemis endonuclease acgvity. The EMBO journal, 25, 3880-3889. Riballo, E., Kuhne, M., Rief, N., Doherty, A., Smith, G.C., Recio, M.J., Reis, C., Dahm, K., Fricke, A., Krempler, A. et al. (2004) A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locagng to gamma-H2AX foci. Molecular cell, 16, 715- 724. 21. Meek, K. (2020) Acgvagon of DNA-PK by hairpinned DNA ends reveals a stepwise mechanism of kinase acgvagon. Nucleic acids research, 48, 9098-9108. 22. 23. Helmink, B.A., Tubbs, A.T., Dorsej, Y., Bednarski, J.J., Walker, L.M., Feng, Z., Sharma, G.G., McKinnon, P.J., Zhang, J., Bassing, C.H. et al. (2011) H2AX prevents CtIP-mediated DNA end resecgon and aberrant repair in G1-phase lymphocytes. Nature, 469, 245-249. Rooney, S., Sekiguchi, J., Zhu, C., Cheng, H.L., Manis, J., Whitlow, S., DeVido, J., Foy, D., Chaudhuri, J., Lombard, D. et al. (2002) Leaky Scid phenotype associated with defecgve V(D)J coding end processing in Artemis-deficient mice. Mol Cell, 10, 1379-1390. 52 24. 25. 26. 27. 28. 29. 30. 31. Roth, D.B., Menetski, J.P., Nakajima, P.B., Bosma, M.J. and Gellert, M. (1992) V(D)J recombinagon: broken DNA molecules with covalently sealed (hairpin) coding ends in scid mouse thymocytes. Cell, 70, 983-991. Liu, L., Chen, X., Li, J., Wang, H., Buehl, C.J., Goff, N.J., Meek, K., Yang, W. and Gellert, M. (2021) Autophosphorylagon transforms DNA-PK from protecgng to processing DNA ends. Molecular cell. Kabotyanski, E.B., Gomelsky, L., Han, J.O., Stamato, T.D. and Roth, D.B. (1998) Double- strand break repair in Ku86- and XRCC4-deficient cells. Nucleic acids research, 26, 5333- 5342. Cui, X. and Meek, K. (2007) Linking double-stranded DNA breaks to the recombinagon acgvagng gene complex directs repair to the nonhomologous end-joining pathway. Proceedings of the Na9onal Academy of Sciences of the United States of America, 104, 17046-17051. Sallmyr, A. and Tomkinson, A.E. (2018) Repair of DNA double-strand breaks by mammalian alternagve end-joining pathways. The Journal of biological chemistry, 293, 10536-10546. Buck, D., Moshous, D., de Chasseval, R., Ma, Y., le Deist, F., Cavazzana-Calvo, M., Fischer, A., Casanova, J.L., Lieber, M.R. and de Villartay, J.P. (2006) Severe combined immunodeficiency and microcephaly in siblings with hypomorphic mutagons in DNA ligase IV. European journal of immunology, 36, 224-235. Chen, S.H. and Yu, X. (2019) Human DNA ligase IV is able to use NAD+ as an alternagve adenylagon donor for DNA ends ligagon. Nucleic acids research, 47, 1321-1334. Dedon, P.C. and Goldberg, I.H. (1992) Free-radical mechanisms involved in the formagon of sequence-dependent bistranded DNA lesions by the angtumor angbiogcs bleomycin, neocarzinostagn, and calicheamicin. Chem Res Toxicol, 5, 311-332. 32. Wu, C.C., Li, T.K., Farh, L., Lin, L.Y., Lin, T.S., Yu, Y.J., Yen, T.J., Chiang, C.W. and Chan, N.L. (2011) Structural basis of type II topoisomerase inhibigon by the angcancer drug etoposide. Science, 333, 459-462. 33. 34. 35. Neal, J.A. and Meek, K. (2019) Deciphering phenotypic variance in different models of DNA-PKcs deficiency. DNA repair, 73, 7-16. Roth, D.B. and Wilson, J.H. (1986) Nonhomologous recombinagon in mammalian cells: role for short sequence homologies in the joining reacgon. Molecular and cellular biology, 6, 4295-4304. Corneo, B., Wendland, R.L., Deriano, L., Cui, X., Klein, I.A., Wong, S.Y., Arnal, S., Holub, A.J., Weller, G.R., Pancake, B.A. et al. (2007) Rag mutagons reveal robust alternagve end joining. Nature, 449, 483-486. 53 36. Oh, S., Harvey, A., Zimbric, J., Wang, Y., Nguyen, T., Jackson, P.J. and Hendrickson, E.A. (2014) DNA ligase III and DNA ligase IV carry out genegcally disgnct forms of end joining in human somagc cells. DNA repair, 21, 97-110. 37. Masani, S., Han, L., Meek, K. and Yu, K. (2016) Redundant funcgon of DNA ligase 1 and 3 in alternagve end-joining during immunoglobulin class switch recombinagon. Proceedings of the Na9onal Academy of Sciences of the United States of America, 113, 1261-1266. 38. Deriano, L., Stracker, T.H., Baker, A., Petrini, J.H. and Roth, D.B. (2009) Roles for NBS1 in alternagve nonhomologous end-joining of V(D)J recombinagon intermediates. Molecular cell, 34, 13-25. 39. Meek, K. (1990) Analysis of Juncgonal Diversity during Lymphocyte-B Development. Science, 250, 820-823. 40. 41. 42. 43. 44. 45. Lewis, S. and Gellert, M. (1989) The mechanism of anggen receptor gene assembly. Cell, 59, 585-588. Normanno, D., Negrel, A., de Melo, A.J., Betzi, S., Meek, K. and Modesg, M. (2017) Mutagonal phospho-mimicry reveals a regulatory role for the XRCC4 and XLF C-terminal tails in modulagng DNA bridging during classical non-homologous end joining. Elife, 6. Kienker, L.J., Shin, E.K. and Meek, K. (2000) Both V(D)J recombinagon and radioresistance require DNA-PK kinase acgvity, though minimal levels suffice for V(D)J recombinagon. Nucleic acids research, 28, 2752-2761. Kurimasa, A., Kumano, S., Boubnov, N.V., Story, M.D., Tung, C.S., Peterson, S.R. and Chen, D.J. (1999) Requirement for the kinase acgvity of human DNA-dependent protein kinase catalygc subunit in DNA strand break rejoining. Molecular and cellular biology, 19, 3877- 3884. Lu, H., Saha, J., Beckmann, P.J., Hendrickson, E.A. and Davis, A.J. (2019) DNA-PKcs promotes chromagn decondensagon to facilitate inigagon of the DNA damage response. Nucleic acids research, 47, 9467-9479. Jiang, W., Crowe, J.L., Liu, X., Nakajima, S., Wang, Y., Li, C., Lee, B.J., Dubois, R.L., Liu, C., Yu, X. et al. (2015) Differengal phosphorylagon of DNA-PKcs regulates the interplay between end-processing and end-ligagon during nonhomologous end-joining. Molecular cell, 58, 172-185. 46. Meek, K., Douglas, P., Cui, X., Ding, Q. and Lees-Miller, S.P. (2007) trans Autophosphorylagon at DNA-dependent protein kinase's two major autophosphorylagon site clusters facilitates end processing but not end joining. Molecular and cellular biology, 27, 3881-3890. 54 47. Roy, S., de Melo, A.J., Xu, Y., Tadi, S.K., Negrel, A., Hendrickson, E., Modesg, M. and Meek, K. (2015) XRCC4/XLF Interacgon Is Variably Required for DNA Repair and Is Not Required for Ligase IV Sgmulagon. Molecular and cellular biology, 35, 3017-3028. 48. Mali, P., Yang, L., Esvelt, K.M., Aach, J., Guell, M., DiCarlo, J.E., Norville, J.E. and Church, G.M. (2013) RNA-guided human genome engineering via Cas9. Science, 339, 823-826. 49. 50. Neal, J.A., Xu, Y., Abe, M., Hendrickson, E. and Meek, K. (2016) Restoragon of ATM Expression in DNA-PKcs-Deficient Cells Inhibits Signal End Joining. Journal of immunology, 196, 3032-3042. Ochi, T., Gu, X. and Blundell, T.L. (2013) Structure of the catalygc region of DNA ligase IV in complex with an Artemis fragment sheds light on double-strand break repair. Structure, 21, 672-679. 55 Chapter 3: New insight into how the DNA binding domain of DNA ligase IV facilitates end- joining, independent of its catalyZc acZvity. By: Noah J. Goff, Saanjh Sundar, Addy Walia, Gargi Parkhi, Katheryn Meek Data Chapter 56 ABSTRACT DNA double strand breaks (DSBs) are highly genotoxic lesions generated throughout the cell cycle by cellular processes (DNA tension, replicagon errors, metabolic stress) or exogenous agents (ionizing radiagon, chemotherapeugc drugs). Cells have several pathways to resolve DSBs and miggate their insult to genomic integrity. End-joining pathways, most notably non-homologous end joining (NHEJ), are the most readily available DSB repair mechanisms—being available throughout the cell cycle and capable of repairing a wide range of damage adducted DNA-ends. Recent studies have revealed a two-stage synapgc mechanism for NHEJ, where DNA Ligase IV (L4) acts both catalygcally to ligate the broken strands and as an essengal structural factor that drives progression from long-range synapgc complexes (LRCs) into a ligagon-capable short-range synapgc complex (SRC). Here we report a novel cryo-EM structure containing density for L4’s DNA binding domain (DBD) in the LRC, something not previously seen in any reported structures. The L4 side of that interface is shared with a trans interacgon between L4 and Ku70 in the SRC. We characterize how the L4 DBD promotes end-joining in the absence of L4 catalygc acgvity through interacgons in the SRC with DNA and Ku70. Each of these interacgons are essengal to the capacity of catalygcally inacgve L4 to promote joining. Moreover, we have discovered that catalygcally inacgve L4 requires a specific LRC to promote end-joining. Finally, we have observed several instances of a single structural interface in one protein mediagng interacgons with different NHEJ components, and we discuss potengal implicagons of this finding. 57 INTRODUCTION Preservagon of genegc material is paramount to long-term health and survival of all organisms. Threats to genomic stability either result from common environmental agents or are byproducts of essengal cellular funcgons, including reacgve byproducts of metabolism, torsional stress within the nucleus, autonomous nuclear elements, and some viruses among other sources. Failure of DNA repair pathways in eukaryotes omen results in harmful lesions that can be converted into mutagons, impair cellular funcgon, contribute to development of cancers, or eventually drive cell death. DNA double strand breaks (DSBs) are considered the most genotoxic lesions—with inappropriate repair resulgng in a range of outcomes from small delegons to large chromosomal rearrangements (79–81, 52, 45). Direct end-joining is the primary repair mechanism available throughout the cell cycle and is primarily mediated through the canonical non-homologous end-joining pathway (NHEJ) (82). A general model of NHEJ begins with sensing damage, synapsis of two ends, processing of chemical adducts to produce compagble 3’ hydroxyl and 5’ phosphate groups and complegon of the process by direct ligagon of the DNA ends. The core NHEJ factors in mammals (and many other eukaryotes)(35) include the DNA dependent protein kinase [DNA-PK, consisgng of the DNA end- binding heterodimer, (Ku70/Ku80), and the catalygc subunit DNA-PKcs] that acts as a damage sensor, structural factors XRCC4, XRCC4-Like Factor (XLF) and paralog of XRCC4 and XLF (PAXX), and the NHEJ-specific DNA ligase IV (L4) that performs the final ligagon step. NHEJ’s availability throughout the cell cycle addresses a central challenge of cellular DNA repair: how to repair lesions when a homologous template is rarely available? As a result, NHEJ is both remarkably flexible and fast, but increases the risk of small indels or larger chromosomal instability. Recent work in the field has developed a more detailed two-stage synapgc model of NHEJ, where ends are synapsed and protected from aberrant end processing in a “long-range” synapgc complex, essengal end processing is performed and required downstream factors are recruited before ulgmately transigoning into a “short-range” complex where ends become accessible to either L4 or further end-processing by X-family DNA polymerases lambda and mu (Poll and Polm), the phosphodiesterase TDP1, and the nucleases Artemis, MRN, and others (21, 69, 83, 84). 58 Work from Loparo and colleagues first proposed a two-stage model of DNA end synapsis by NHEJ factors (46). Their single molecule FRET studies revealed both short-range and long-range synapses where ends are held in either direct proximity (short-range complex) or ~115Å apart (long-range complex) termed SRC and LRC respecgvely. Transigon into a short-range complex required the presence of Ku70, Ku80, XLF, XRCC4, L4, DNA-PKcs kinase acgvity (but not necessarily its presence), and crigcally L4, but not its catalygc acgvity (46). Further FRET and biochemical studies from these authors revealed specific roles for Poll mediated fill-in synthesis and TDP1- dependent phosphodiesterase in the SR complex (21). Interesgngly, while Loparo and colleagues worked on single molecule imaging, new structural data from several groups revealed diverse NHEJ complexes consisgng of DNA-PK bound to a single DNA end or in complex with other NHEJ factors (XLF, XRCC4/L4, PAXX) to tether two ends together in a dimeric structure. Remarkably, these new NHEJ structures revealed dimeric complexes with DNA ends synapsed in direct contact, in good agreement with the SR complex predicted by the smFRET approaches, as well as mulgple disgnct iteragons of LR synapgc complexes, posigoning the two ends ~115Å apart (18, 22, 70). DNA ends synapsed by the LR complexes can be classified into two groups: 1) Ku80- mediated dimers dominated by trans interacgons between DNA-PKcs and the extreme C-terminal tail of Ku80, and 2) XLF-mediated dimers dominated by a pair of DNA-PKcs interfaces interacgng reciprocally in trans along with a centrally posigoned XLF dimer interacgng with two dimers of XRCC4 (XRCC4-XLF-XRCC4) providing mulgple, protein-protein interfaces that stabilize the DNA synapse. Our recent structure-funcgon mutagenesis studies suggest that LR dimer complexes have disgnct funcgons in promogng NHEJ. Specifically, the Ku80-mediated dimer promotes access to DNA ends for nucleolygc end processing, while the XLF-mediated dimer limits nucleolygc processing—matching the “end protecgon” complex suggested by Loparo and colleagues (20). Our current limited understanding of how NHEJ progresses stems from all of these studies (with all the unique approaches uglized); acgve areas of research now focus on understanding how LR complexes translate between the two dimer forms and what is required to progress from LR complexes to SR complexes. 59 It is now well-appreciated that L4 has an essengal role in promogng the transigon to the short-range complex (10, 21, 46, 73). Recent studies have provided more informagon as to how L4 contributes to the transigon from long-range to short-range end synapsis (20, 70). This funcgon is unique among mammalian DNA ligases. The catalygc domains of the three human DNA ligases contain three mogfs: a DNA binding domain (DBD), a nucleogdyltransferase domain (NTD), and an oligonucleogde oligosaccharide fold domain (OBD). DNA ligases III and VI, but not ligase I (L1, L3, and L4) also include BRCT repeats (39). In the case of L4, a small region of the polypepgde between its two BRCT domains mediates L4’s interacgon with its obligate co-factor XRCC4 (9). L4’s catalygc core has generally not been observed in most of the recent Cryo-EM structures, with the notable excepgon of the SR complex when the catalygc domain is seen bound to a DSB (18– 20). Of note, single molecule imaging experiments revealed that L4 dynamically interacts with each DNA end independently in the LRC; these interacgons are mediated by posigvely charged residues in the DBD that were observed (in Cryo-EM structures of human L4 in the SRC) to make electrostagc interacgons with the DNA backbone, slightly internal to the DNA termini (48). Cortes and colleagues using engineered chimeras of human DNA ligases revealed that chimeric proteins including the BRCT domain of L4 with L1’s NTD and OBD, but not L1’s DBD produces a fusion protein that supports NHEJ-specific VDJ recombinagon (85). The finding that fusing L1’s engre catalygc core (including its DBD) with L4’s BRCT-mogfs was insufficient to promote ligagon via NHEJ, directly implicates L4’s DBD in facilitagng L4's non-catalygc funcgon in NHEJ. More experiments that promote the conclusion that L4 has an important structural role in joining comes from our recent single molecule imaging study showing that whereas L4 is required for stable recruitment of DNA-PK to chromagn amer DNA damage, catalygcally inacgve L4 supports its recruitment. Of note, retengon of DNA-PK at DSBs in cells expressing catalygcally inacgve L4 is markedly longer than acgve L4; these data suggest that repair facilitated by catalygcally inacgve L4 is slower than when L4 is acgve (73). In this study, we further invesggate L4’s structural role in promogng end-joining using NHEJ funcgonal assays in genegcally engineered cell strains and cryo-electron microscopy (Cryo- EM, data not shown). We confirm that repair facilitated by catalygcally inacgve L4 is much slower than with wild type L4, and that repair is pargally dependent on DNA polymerase theta (Polθ). 60 Moreover, repair promoted by inacgve L4 is highly dependent on the Ku80-mediated LRC. In addigon, we report a new version of the Ku80-mediated LRC with density for L4’s DBD interacgng with DNA-PKcs. Of note, the interface that mediates L4's interacgon with DNA-PKcs also facilitates a previously reported trans interacgon between L4's DBD and Ku70 that tethers DNA ends in the SR complex (18). Funcgonal assays establish the relevance of this L4 DBD/Ku70 interacgon for end joining, whereas the interacgon of L4's DBD with DNA-PKcs appears to be dispensable for NHEJ. This interface is disgnct from the two DNA/protein interfaces studied by Sgnson et al (48) that impact L4's tethering of DNA ends in single molecule FRET experiments. We posit that numerous interacgons of the DBD with Ku and the two DNA termini are essengal in promogng transigon from LR to SR complexes where end-joining occurs. A RESULTS B CCTGCAG GGACGTCTTAAG CGCGTCCTGCAG AGGACGTC Figure 13: Microhomology mediated end joining is mediated through Polθ independent of NHEJ deficiency. A) Schemagc of MMEJ fluorescent reporter assay, with 7 bp of microhomology highlighted and underlined. Substrates were pre-cut with ApoI and MluI (NEB) to generate noncomplementary overhangs, then cotransfected with a ECFP plasmid to assess transfecgon efficiency. Use of this microhomology is required to restore the full ds-red reading frame to allow MMEJ+ signal. MMEJ+ signal was assessed as % of transfected cells that expressed DsRed. B) MMEJ joining acgvity in edited 293T cell strains, independent of L4 presence or catalygc acgvity. Stagsgcs performed using an unpaired two-tailed Student’s t-test (**** p > 0.0001). 61 Microhomology-mediated end joining (MMEJ) is dependent on Polθ. We recently showed that catalygcally inacgve L4 promotes substangal end-joining and radioresistance in cell culture models, bolstering earlier reports documengng a non-catalygc role for L4 in NHEJ. Our study suggests that joining in cells expressing inacgve L4 [a K273A mutagon] is likely mediated by L3, but in the context of an undefined NHEJ complex (10). This cooperagon between L4 and L3 is strongly corroborated by a recent collaboragve study of catalygcally inacgve L4 in mice that lack nuclear L3 (14). In a separate publicagon, analyses of NHEJ factor recruitment to DNA DSBs in living cells revealed that whereas in cells lacking L4, NHEJ factor recruitment (using Halo-Flag tagged Ku70, DNA-PKcs, and XRCC4) is transient (in the case of Ku70 and DNA-PKcs) and absent for XRCC4; but in cells expressing the K273A mutant, although inigal NHEJ factor recruitment occurs similarly as in cells expressing acgve L4, dissociagon of NHEJ factors is markedly stalled (73). The increased retengon of NHEJ factors in cells expressing L4 K273A suggests that repair may occur less rapidly than in NHEJ-proficient cells where end-joining is rapid, and (when DNA ends are ligatable and compagble) generally accurate. Sequencing across rejoined DSBs in cells expressing catalygcally inacgve L4 revealed lower fidelity repair outcomes with an increased dependence on ligagng ends across short homologous sequences near the break (1-5 bp, frequently referred to as microhomology). This is characterisgc of end-joining facilitated by Polθ, which is notably slower than NHEJ (and therefore only happens at low levels in NHEJ proficient cells) . Addigonally, rejoined DSBs from cells expressing L4 K273A share characterisgcs of joints mediated by Polθ (increased nucleogde loss and increased use of terminal microhomology) (15). To assess the role of Polθ in cells expressing L4 K273A, Polθ was ablated using a CRISPR/Cas9 strategy; the impact of Polθ loss specifically on repair of DSBs via terminal microhomology was tested using a linearized plasmid substrate that requires use of a 7-bp short-sequence homology internal to the break site to restore an RFP open reading frame. All cells proficient in Polθ, regardless of NHEJ proficiency, support basal levels of MMEJ. In contrast, MMEJ joining is remarkably reduced in Polθ ablated cells expressing either wild-type L4 or catalygcally inacgve L4 (Figure 13B). [This joining substrate reports only on MMEJ mediated joints, explaining the independence from NHEJ acgvity.] Consistent with previous reports (77, 86, 87), Polθ mediated 62 end joining funcgons independently from end joining catalyzed by NHEJ. We conclude that Polθ is required for MMEJ mediated joining, and that catalygcally inacgve L4 cannot facilitate MMEJ in the absence of Polθ. A B C Figure 14: CatalyZcally inacZve L4 supports end joining through mulZple alt-EJ pathways. A) Sensigvity of 293T cells to teniposide as assessed by MTT assay. B) Sensigvity of 293T cells to calicheamicin as assessed by MTT assay. C) Time-course version of a VDJ episomal assay, with fixed cells represengng joining amer 24, 48, or 72 hours as analyzed by flow cytometry. In cells expressing catalyZcally inacZve L4, resistance to DSB-inducing agents requires Polθ. Since Polθ promotes substangal amounts of MMEJ, we reasoned that MMEJ may play a more prominent role in the alternagve end joining promoted by cells expressing L4 K273A. Cells deficient in Polθ were not sensigve to either calicheamicin (that generates DSBs with 3’ overhangs with 3’ phosphoglycolate adducts) or teniposide (a type II topoisomerase poison that leaves 5’- pepgde adducted DSB overhangs primarily in S phase). However, L4 K273A expressing cells that 63 lack Polθ, are markedly sensigve to both drugs, approaching the level of sensigvity observed in cells that completely lack L4 (Figure 14A-B). Because Polθ cannot facilitate sufficient end-joining in L4 deficient cells (as evidenced by a synthegc lethal interacgon between loss of Polθ and NHEJ), these data suggest that Polθ may promote end-joining in the context of a catalygcally inacgve L4 complex. VDJ coding joining facilitated by catalyZcally inacZve L4 is slower than wild type NHEJ and is not dependent on Polθ. Time course assays were performed to assess the rate of VDJ coding end joining (Figure 14C). As shown previously, cells expressing K273A L4 join RAG-induced DSBs (in this case coding ends) much more efficiently than cells lacking L4 (10). However, joining in the K273A cells is delayed compared to joining from cells expressing acgve L4. Finally coding joint efficiency or rate is not impacted by loss of Polθ in cells expressing either wild type or K273A L4. Altogether, these data suggest that K273A L4 contributes to a variety of repair mechanisms, including both Polθ mediated alt-EJ and L3-mediated joining [as shown by our recent collaboragve study (14)]. End-joining facilitated by K273A L4 requires the Ku80-mediated long-range NHEJ complex. As noted above, accumulagng data support a new model whereby NHEJ funcgons by inigally synapsing two DNA ends in LR complexes posigoning the ends ~115 Å apart. A fracgon of LRCs transigon to SR complexes that juxtapose ends close enough to facilitate ligagon (46). Emerging cryo-EM data have dramagcally bolstered and expanded this model providing structural evidence for a variety of disgnct LRCs, but only one SRC (18–20). What is lacking is a clear understanding of how complexes transigon from one form to another, or if catalygcally inacgve L4 is dependent on a specific complex. The slow rate of joining in cells expressing K273A L4 as well as the delayed dissociagon of NHEJ factors from chromagn in cells expressing K273A L4 might suggest stalling in a pargcular complex. To test the impact of L4 inacgvity when DNA-PK mutagon disrupts normal transigon between LR and/or SR complexes, a series of episomal end-joining assays were performed in a 293T K273A cells strain that was ablated for DNA-PKcs expression using a CRISPR strategy. In these assays, the interplay between DNA-PKcs and L4 can be ascertained by complemengng the DNA- PKcs deficiency using mutant constructs that disrupt either the Ku80-mediated dimer (4A) or the 64 XLF-dependent dimer (898/2569) or both (4A/2569, Figure 15). In these experiments VDJ coding and signal joining assays were performed using a previously described hyper-RAG2 mutant that induces substangally higher levels of recombinagon; this accentuates differences in end-joining capacity. As can be seen, K273A cells support substangal VDJ coding and signal end-joining in cells complemented with wild type DNA-PKcs (although sgll less than observed in cells with wild type L4). As reported previously, DNA-PKcs mutants disrupgng either of the two LR complexes have modest deficiencies in joining in cells with wild type L4 (20). When complemengng the DNA-PKcs deficiency with the 898/2569 mutant that disrupts the XLF-dependent dimer, signal and coding end-joining are proporgonally reduced in cells with K273A L4, as compared to cells with wild type L4. In contrast, both signal and coding end-joining in cells expressing inacgve L4 is virtually ablated in complementagon experiments with DNA-PKcs mutants that disrupt the Ku80-mediated dimer (4A and 4A/2569, Figure 15). These data suggest that interacgons mediated by the domain swap dimer are required to facilitate joining promoted by inacgve L4. Figure 15: End-joining facilitated by K273A L4 requires the Ku80-mediated long-range NHEJ complex. A-B) VDJ coding end (lem) and signal end (right) joining assays in 293T PKcs-/- L4+/+ (solid bars) and PKcs-/- L4K273A/- (striped bars). L4’s DNA binding domain (DBD) interacts with the M-HEAT region of DNA-PKcs in the Ku80- mediated dimer. During refinement of recent NHEJ complexes, a density previously not appreciated was idengfied as a part or L4’s DBD (Chloe L. Hall, Steven W. Hardwick, and Amanda K. Chaplin. Data not shown). Of note, L4's DBD has not previously been observed in any of the 65 different forms of the LRC (18, 19). This interacgon appears to be mediated by two highly conserved posigvely charged residues R136 and K140. L4 R136 interacts with D1440 in DNA-PKcs whereas L4 K140 interacts with DNA-PKcs at residues 1495-1498. There is also a potengal interacgon of a loop within the FAT domain (3197-3226) that interacts with L4's acgve site. Finally, there is a potengal interacgon of L4 K264 with Ku70 T307. This density for L4 and its interacgon with DNA-PKcs has only been observed in the Ku80-mediated dimer. The DNA-PKcs interface that interacts with L4’s DBD is not required for efficient NHEJ. To ascertain the funcgonal relevance of this DNA-PKcs/L4 interacgon, expression constructs ablagng the interacgng interfaces in the two proteins were generated. In DNA-PKcs, residues D1440 and E1497 were changed to arginine and lysine respecgvely (DE>RK). In L4, residues R136 and K140 in the DBD were subsgtuted with aspargc acid (RK>DD). Mutant DE>RK DNA-PKcs was tested in transient episomal end-joining assays in the DNA-PKcs deficient 293T cell line described above. As can be seen, DNA-PKcs DE>RK restores end-joining at or above WT levels (in this case VDJ coding joints) as well as wild type DNA-PKcs (Figure 16B). Because we considered that the tethering of L4 might be relevant to the LR to SR transigon (that is possibly altered in cells with inacgve L4), end-joining was also tested in the K273A expressing 293T cells with CRISPR-ablated DNA-PKcs (described above, Figure 16C). DE>RK mutant DNA-PKcs restores joining to a similar level as wild type DNA-PKcs, albeit at a reduced level compared to cells expressing acgve 293T cells. We next generated stable cell strains expressing either wild type, DE>RK, or no DNA-PKcs in the V3 CHO cell strain that lacks DNA-PKcs (Figure 16D). While cells lacking DNA-PKcs are hyper- sensigve to both calicheamicin and teniposide, cells expressing DE>RK mutant DNA-PKcs are similarly resistant to these DSB-inducing agents as wild type DNA-PKcs. We conclude that the interface in DNA-PKcs that interacts with L4’s DBD in the Ku80-mediated dimer is not required for cellular resistance to DSB-inducing drugs or for rejoining DSBs in episomal end-joining assays. 66 Figure 16: The DNA-PKcs interface that interacts with L4’s DBD is not required for efficient NHEJ. A) 293T Western with transient complementagon of Vect, WT, DE>RK in both Lig4+/+ and Lig4K273A*/- B) 293T joining assays, VDJ coding PK22 lem c195-14 right C) Western assessing DNA- PKcs expression in clonal populagons of CHO-V3 cells complemented with empty vector (vect), WT DNA-PKcs (WT) or DNA-PKcs DE>RK (DE>RK) D) Colony formagon assays tesgng sensigvity to Calicheamicin (lem) and teniposide (right). The L4 interface that interacts with DNA-PKcs is important for efficient NHEJ in cells with acZve L4, but essenZal for NHEJ in catalyZcally inacZve L4. Expression vectors encoding L4 with only the RK>DD subsgtugons were prepared; in addigon, the RK>DD subsgtugon was combined with addigonal L4 mutagons targegng its catalygc acgvity [including the K273A mutagon that ablates the major adenylagon site, and also K273A with five addigonal lysine>arginine subsgtugons (5xK) that ablate a small level of “back-up” adenylagon observed in in vitro experiments with the K273A mutant] (10). To test whether the DBD interface that can interact with DNA-PKcs impacts joining, episomal end-joining assays as described above were performed. Consistent with our previous study, in the absence of L4, virtually no VDJ coding end joining is observed (Figure 17A-C); in contrast, wild type L4 (WT), as well as catalygcally inacgve L4 mutants (K273A, 5xK mutants) all facilitate substangal coding end joining. When L4 is acgve, the RK>DD mutant promotes substangal coding end joining, albeit reduced compared to acgve L4. However, when the RK>DD mutagon is combined with inacgve L4, joining is virtually ablated. 67 It is well-established that the RAG1/2 endonuclease directs its DSBs into the NHEJ pathway by a poorly understood mechanism (reference). To address whether the RK>DD mutant impacts joining of DSBs that are not exclusively repaired by NHEJ, joining of DSBs induced by both TelN (a DNA hairpin forming phage protelomerase) and I-SceI (an endonuclease that produces noncomplementary, 4 base pair 3’ overhangs) were studied. We previously found that cells expressing inacgve L4 can join non-RAG induced DSBs more efficiently than RAG DSBs (10). Similar to VDJ-coding hairpins, cells expressing the RK>DD mutant are modestly deficient in joining TelN induced DSBs; the deficiency is exacerbated when the RK>DD subsgtugon is combined with loss of catalygc acgvity (Figure 17D). Interesgngly, cells expressing the mutant combining inacgve L4 with the RK>DD subsgtugon are also severely deficient in joining non-hairpin I-SceI breaks despite the increased level of joining reported previously with inacgve L4 (12). Stable complementagon of wild type and mutant L4 was performed uglizing a PiggyBac retrotransposon system to stably introduce L4 into L4 deficient 293T and U2OS cell lines. To test whether cells expressing the RK>DD mutant are also more sensigve to agents that induce genomic DSBs, stable cell strains expressing wild type or mutant L4 were treated with calicheamicin and teniposide. Cells expressing inacgve L4 (either K273A or 5xK) or the RK>DD mutant L4 are more resistant to calicheamicin and teniposide than cells lacking L4. Consistent with the end-joining assays, L4 mutants that combine the RK>DD subsgtugon with mutagons that disrupt catalygc acgvity are as sensigve to calicheamicin as cells lacking L4 (Figure 17B). We conclude that R136 and K140 are crigcal for end-joining; the observagon that the impact of mutagon of these residues potengates deficits in joining mediated by catalygcally inacgve L4 suggests that interacgons mediated by these residues are important for L4's non-catalygc role in repair via NHEJ. 68 A D G B C E2-Crimson ZsGreen RAG/TelN/I-SceI Cut Sites E F H I D D > K R + A 3 7 2 K D D > K R + K x 5 K x 5 D D > K R A 2 7 2 K t c e V T W DNA-PKcs Lig4 Figure 17: The L4 interface that interacts with DNA-PKcs is important for efficient NHEJ in acZve L4, but essenZal for NHEJ in catalyZcally inacZve L4. A) Diagram of the joining assay substrate, with cut sites . Substrates were cotransfected into cells along with and without enzyme (+/-) and appropriate L4 expression vector, then red/green fluorescence via flow cytometry at 72 hours. B) Results from the VDJ coding- and C) signal-end substrates when cotransfected with Rag1 and hyper mutant Rag2 along with the corresponding L4 expression vector. D-E) TelN/I-SceI substrates cotransfected with endonuclease (-) or with appropriate endonuclease and corresponding L4 expression vector. F) VDJ coding-end joining assays as before tesgng addigonal mutagons in the L4 DBD G) Sensigvity of 293T L4-/- cells complemented with PiggyBacc-expressed L4 mutants to calicheamicin as assessed by MTT assay, compared to 293T L4+/+ (termed 293T WT). H) Sensigvity of PiggyBacc-edited U2OS L4 cell strains as assessed by clonogenic survival assays. I) Immunoblot showing transient L4 expression in a 293T L4 -/- cell strain with DNA-PKcs levels as a control. The L4 interface that interacts with DNA-PKcs in the Ku80-mediated dimer forms a highly conserved salt-bridge interacZon with Ku70 in the short-range complex. Mulgple Sequence 69 Alignments performed by COBALT revealed substangal conservagon of R136 and K140, from numerous vertebrate species (Figure 18). To clarify the discrepancies of mutagng these two interacgng interfaces in L4 and DNA-PKcs, we searched published structures (18, 19, 22) of mulgmeric NHEJ complexes for interacgons between L4 and structural factors that could explain how disrupgon of R136 and K140 might impact NHEJ. R136 and K140 are only structured in two available structures of NHEJ complexes: the interacgon with DNA-PKcs described above, and in the short-range complex reported by He and colleagues (18). In this publicagon, L4 residues 134- 143 were observed interacgng with the von Willebrand domain of Ku70. Specifically, R136 in L4's DBD bridges directly with D192 in Ku70 in a trans interacgon (Figure 17). We posit that L4's ability to tether two DNA ends is mediated by numerous interacgons including 1) protein/DNA interacgons involving two patches of posigvely charged residues described by Loparo and colleagues (48), 2) protein/DNA interacgons with L4's catalygc site, 3) the trans interacgon between L4 R136 and K140 with Ku70 D192 (18), and finally 4) the capacity to form the Ku80- mediated dimer. Whereas disrupgon of any one of these interacgons results in only a pargal loss of end-joining and radio-resistance, loss of two results in severe deficits in both. 70 B D F Homo sapiens Mus musculus Ra>us norvegicus Danio rerio Xenopus laevis Equus caballus Gallus gallus Bos taurus Canus lupus familiaris Pan troglodytes Homo sapiens Mus musculus Ra>us norvegicus Danio rerio Xenopus laevis Equus caballus Gallus gallus Bos taurus Canus lupus familiaris Pan troglodytes Homo sapiens Mus musculus Ra>us norvegicus Danio rerio Xenopus laevis Equus caballus Gallus gallus Bos taurus Canus lupus familiaris Pan troglodytes Homo sapiens Mus musculus Ra>us norvegicus Danio rerio Xenopus laevis Equus caballus Gallus gallus Bos taurus Canus lupus familiaris Pan troglodytes A C E G I H Homo sapiens Mus musculus Ra>us norvegicus Danio rerio Xenopus laevis Equus caballus Gallus gallus Bos taurus Canus lupus familiaris Pan troglodytes Homo sapiens Mus musculus Ra>us norvegicus Danio rerio Xenopus laevis Equus caballus Gallus gallus Bos taurus Canus lupus familiaris Pan troglodytes Figure 18: IdenZfying conserved shared interfaces between NHEJ factors. A) Cryo-EM structure of the Short-Range NHEJ dimer (PDB: 7LSY) highlighgng an interface between the ligagon- posigoned Lig4 molecule (green) and Ku70 (black). Lig4 residues R136 and K140 are highlighted in magenta and form the basic side of a salt bridge with Ku70. Also pictured Ku80 (black), … 71 Figure 18 (cont’d) XRCC4 (Purple), XLF (pink), and the BRCT-structural domain of a second Lig4 molecule (gold). B) Species conservagon of Lig4 orthologs in 10 vertebrate species aligned to human Lig4 K134–L143. C) Cryo-EM structure of the XLF-dependent DNA-PK long-range NHEJ dimer highlighgng an interacgon between Lig4 and Ku70/Ku80. Zoomed in diagram showing Lig4 N692, R708, N711 (magenta) interacgng with Ku80. D) Species conservagon of Lig4 orthologs in 10 vertebrate species aligned to human Lig4 K134–L143. E) Shared interface between Ku80 and Lig4-BRCT in the Ku80-mediated dimer with PAXX/XRCC4/L4 (PDB 8BHY, Lig4 N711 highlighted in magenta). F) Species conservagon of Ku70, with D192-D196 highlighted in red. G) Species conservagon of Ku80 D327-Q330 ”DEEQ mogf” (red) aligned to human Ku80. H) Conservagon of DNA-PKcs D1440 (red). I) Conservagon of DNA-PKcs E1497 (red). DNA-interacZng residues in the L4 DBD work cooperaZvely with the Ku70 trans interface to join ends. A recent study by Loparo and colleagues revealed that L4 dynamically interacts with ends prior to SRC synapsis, mediated by 4 basic residues in its DBD that directly interact with DNA in cryo-EM structures. Mutagng these interacgons significantly impaired interacgons with DNA and disrupted stable SRC formagon. We hypothesized that combining the DNA-interface mutant with catalygcally inacgve L4 would impair its ability to promote end joining, similar to K273A + the RK>DD mutagon targegng the L4 DBD-Ku70 interface. To address this, we designed an expression vector mutagng the equivalent residues in human L4 (K28E, K30E, R32E, K162E, R163E, K164E, together termed mDBD) and tested the ability to join VDJ coding-end substrates (Figure 17F). In cells expressing catalygcally acgve L4, mDBD decreases joining by ~50%, indicagng that disrupgng DNA-binding significantly (but not completely) impacts the ability of NHEJ to promote joining. Combining mDBD with either RK>DD or L4 K273A completely ablates all joining. This suggests that 1) ligagon via L4 is structurally supported both by protein-DNA interacgons and protein-protein interacgons between L4’s DBD and Ku70’s VWA, and 2) that the alternagve end-joining sgmulated by catalygcally inacgve L4 is dependent on its structural interacgons that stabilize the SRC. DISCUSSION A single protein interface may have mulZple funcZons, potenZally supporZng flexibility in complex formaZon or driving transiZons between disZnct complexes. Here we report funcgonal significance of a L4 protein-protein interacgon recently idengfied in a new form of the Ku80 mediated LRC. To address the funcgonal relevance of this interacgon, a mutagonal approach was 72 uglized to ablate the interface by subsgtugng residues in both DNA-PKcs and L4. In contrast to many of our previous studies where mutagon of either side of a single interface generally results in similar cellular phenotypes, mutagons designed to ablate either the L4 or DNA-PKcs side of the interface had very different cellular impacts. Of note, ablagng the L4 surface impacted funcgon, and this impact was enhanced when L4’s catalygc acgvity was also blocked. Amer examining published structures of NHEJ complexes, we observed that the L4 residues involved (R136/K140) also mediate a trans interacgon with Ku70 in the NHEJ short-range complex. This study suggests a model where the L4 interface that interacts with DNA-PKcs in the Ku80-mediated dimer may eventually transit (either directly or indirectly) into a more consequengal interacgon with Ku70 that we posit contributes to tethering DNA ends prior to ligagon. Of note, we have observed a similar dual funcgon of another interface in DNA-PKcs. The four highly conserved lysine residues in the M-HEAT region of DNA-PKcs that facilitate both cis and trans interacgons with Ku80's C-terminal acidic pepgde (20), also interact with a disgnct interface in the recently described LR-ATP complex reported by He and colleagues that likely represents a transigonal LR complex (54). Finally, we have recently observed via cryo-EM experiments, Ku-bound to chromagnized DSBs (unpublished data, Amanda Chaplin). In this structure, we observe a specific interface in Ku80 that interacts with the H3 nucleosome subunit. However, this same interface in Ku80 also mediates its interacgon with the BRCT domains of L4 observed in many NHEJ complexes (both long- and short-range). One explanagon for these observagons is that single interfaces that facilitate assembly of different complexes may dictate how transigons between NHEJ complexes proceed. Lig4 plays an essenZal structural role in tethering DNA ends prior to ligaZon. Single molecule imaging studies from Loparo and colleagues demonstrate that interacgons of L4's DBD are crigcal for progression from long-range to short-range complexes (48). More specifically, crigcal DNA- interacgng residues in the L4's DBD dramagcally reduced L4-DNA interacgons in the LR complex and subsequent SR complex formagon, indicagng that this noncatalygc interacgon between L4 and the break serves as an essengal step preceding SR synapsis. Their data suggests a model where L4 dynamically interacts with ends protected by DNA-PKcs, and L4's binding of the two ends promotes progression to the SR complex. Our recent report established that catalygcally 73 inacgve L4 delays release of Ku, DNA-PKcs, and XRCC4 from chromagn amer DNA damage in living cells (73); from these data, we posit that the catalygc site of L4 also promotes progression to short-range complexes. The descripgon here of another L4 DBD interacgon (R136 and K140 in L4 with D192 in Ku70) that funcgons collaboragvely with the catalygc site may also impact L4's ability to promote progression to short-range complexes underscores the central role of L4 in this transigon. These data prompted an examinagon of the same L4/DNA end interacgon in the cellular assays used in this study. Altogether, these studies suggest that L4 may serve as a molecular “sensor” in the long-range complex, promogng transigon into the ligagon-competent short-range complex only if both DNA ends are appropriate substrates for ligagon. This "sensor" funcgon is dependent not only on L4's interacgon with DNA ends, but also L4's interacgon with Ku70, and L4's catalygc site. L4 is unique among eukaryogc ligases in its characterisgc as a single-turnover enzyme (51); thus, it is quite possible that the L4 molecule that seals one strand does not necessarily seal the second strand. One possibility is that rapid ligagon of one strand and NHEJ-complex mediated end-tethering act cooperagvely to support ends. Conversion of a DSB to a single strand break through one-step ligagon would dramagcally reduce the threat of the lesion and the conversion may allow NHEJ factors to “make way” for other DNA repair pathways to resolve the second strand’s break. In sum, these data underscore how the molecular dynamics of NHEJ complexes are highly dependent on L4 and its structural interacgons. InacZve L4 requires the Ku80-mediated dimer for funcZon. We have shown previously that the two forms of long-range synapgc complexes have disgnct funcgons and are in equilibrium. The observagon that cells expressing K273A L4 have prolonged DSB-induced chromagn associagon, suggested to us that long-range to long-range or long-range to short-range transigons might be impacted by loss of L4's catalygc acgvity. This prompted an examinagon of whether either of the two LR complexes are funcgonally crigcal in cells expressing K273A L4. Our data demonstrate show that the Ku80-mediated dimer is essengal for K273A L4 to promote end-joining, and we suggest that K273A L4 promotes alternagve end-joining in the context of the Ku80 mediated dimer. 74 L4's essenZal non-catalyZc role that facilitates end-joining is mediated by disZnct interacZons in NHEJ short-range complexes and involves disZnct alternaZve joining pathways. NHEJ complexes in living cells expressing K273A L4 have significantly different characterisgcs than cells expressing wild type L4. NHEJ factors in complex with K273A L4 display prolonged associagon with chromagn in response to damage, suggesgng that although catalygcally inacgve L4 facilitates NHEJ complex formagon, progression of those complexes is delayed (73). Clearly, L4’s structural interacgons with other NHEJ factors, and not just its catalygc interacgons with DNA are crigcal for promogng end-joining. But how does catalygcally inacgve L4 promote joining? The data presented here and previously demonstrate that disgnct end- joining mechanisms are facilitated by the presence of inacgve L4. We have shown previously that K273A robustly facilitates L3 mediated joining in vitro; these data are strongly corroborated by our recent collaboragve study from Yu and colleagues demonstragng a synthegc lethal interacgon between catalygcally inacgve L4 and loss of nuclear L3 in mice (14). Here we show that unlike loss of L4, inacgvagon of L4 is not synthegcally lethal with Polq ablagon. Moreover, in K273A L4 expressing cells, whereas VDJ joining is minimally impacted by loss of Polq, both MMEJ and resistance to DSB-inducing agents are strongly impacted by loss of Polq. From drug sensigvity and episomal joining assays, we show that alternagve end-joining sgmulated by catalygcally inacgve L4 is dependent on stable promogon of the SRC. Furthermore, since 293T L4K273A/- Polθ-/- and cells expressing L4 K273A + RK>DD are comparably sensigve to DSB inducing agents, we can infer that the high degree of SRC stability with catalygcally inacgve L4 reported by mulgple groups (73) is responsible for promogng alt-EJ in the context of catalygcally inacgve L4. An emerging consensus in the end-joining field is that there are mulgple alternagve end-joining pathways. The data presented here suggest that catalygcally inacgve L4 promotes repair by several of these pathways, likely thorough its ability to promote sustained end-processing in the SRC. Conclusions: All together, these studies further bolster a perhaps incontrovergble conclusion that L4 has mulgple disgnct structural roles in NHEJ. What is lacking is an understanding of 1) the molecular mechanism of transigon between the two forms of long-range complexes, 2) the transigon from long- to short-range complexes and 3) the stoichiometry of DNA ligases in the short-range complex—pargcularly for difficult ends that are not rapidly ligated. 75 MATERIALS AND METHODS Cell culture and genome ediZng 293T and U2OS cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM, Life Technologies) and Roswell Park Memorial Insgtute 1640 Medium (RPMI), respecgvely, supplemented with 10% fetal bovine serum (Atlanta Biologicals, GA), 2 mM L-glutamine, 0.1 mM non-essengal amino acids, 1 mM sodium pyruvate, 100 U/ml penicillin, 100 g/ml streptomycin and 25 ng/ml Gibco Amphotericin B (Life Technologies). TransfecZons of 293T and U2OS cell lines were performed using unsupplemented RPMI media, polyethyleneimine (PEI, 1 µg/mL Polysciences), and plasmid DNA at a rago of 100 µL RPMI:1 µg DNA:2 µg PEI. First, plasmid DNA and RPMI were well-mixed in a 1.5 mL microcentrifuge tube followed by addigon of PEI. Transfecgon mixes were shaken briefly to thoroughly mix contents, then incubated at room-temperature for up to 30 minutes to allow PEI:DNA complexes to form before adding the reacgon volume to freshly plated cells Genome ediZng PolQ delegon in 293T cell strains was performed via CRISPR/Cas9 by targegng cells with a pair of gRNAs spaced 2.1 kb apart in the polq locus to create a large delegon. Each gRNA was cloned into a pCas-2A-Puro (Addgene 62988), then 1 µg of each plasmid was transfected into a 293T L4- /- and L4K273A/- cell strains. Clonal populagons were grown out for one week and screened via outside-outside PCR (to idengfy clones with large delegons at the polq locus). Candidates were then checked for presence of remaining WT allele. pPB stable L4 complementagon: Transfected 1.0E6 cells with 1.5 µg pPB-Puro-Lig4 payload and 1.5 µg pBase transposase plasmids. Cells were allowed to recover for 48 hours before selecgon with 1-2 µg/mL puromycin. Bulk populagons were cultured in the presence of puromycin for the duragon of the experiments. Cell Survival assays: U2OS L4-/- cell strains complemented with PiggyBacc L4 mutants were assessed for resistance to DSB inducing drugs using colony formagon assays. Briefly, 200-300 cells of each mutant strain were plated into 6 cm gssue culture dishes with increasing concentragons of drug and allowed to grow for 9 days. Plates were stained with 2% crystal violet solugon (VWR) and counted. A minimum of 3 Replicates was performed for each strain at each concentragon. 293T+L4 mutant cell strains were assessed using MTT-metabolic assays as a 76 marker for cell viability. 1.6E4 cells were plated in wells of a 24-well plate at increasing concentragons of calicheamicin in 1 mL of DMEM media. Amer 4 days of drug treatment, cells were treated with 1 mg/mL MTT (Goldbio) for 1 hr at standard culture condigons. MTT was removed and cells were dissolved in 200 µL 100% DMSO and absorbance was sampled on a plate reader at 570 nm wavelength. A well without cells treated with media + MTT was used as a background control subtracted from all absorbance values. Episomal Joining Assays: 293T L4-/- cells were transfected following our PEI protocol with a mixture of previously reported episomal joining substrates (10, 11, 88, 89), endonucleases, and plasmid expression vectors encoding L4 mutants in a 1:1:1 rago (by plasmid mass), then cultured for 72 hours before paraformaldehyde fixagon before running on an Ajune CytPix flow cytometer. MMEJ assays were similarly transfected into 293T cells with 1.0 µg linearized DNA substrate and 0.1 µg pECFP transfecgon marker, with 2.0 µg PEI, 100 µL RPMI and fluorescence analyzed 72 hours later gagng for CFP+ transfected cells, with the subpopulagon of dsRed+ cells represengng MMEJ+ outcomes. ACKNOWLEDGMENTS We would like to thank Daniel Vocelle and the MSU Flow Cytometry core for operagng and maintaining the Ajune CytPix, supported by the Equipment Grants Program, award #2022- 70410-38419, from the U.S. Department of Agriculture (USDA), Nagonal Insgtute of Food and Agriculture (NIFA). 77 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. REFERENCES Waters,C.A., Nonhomologous end joining: A good solugon for bad ends. DNA Repair, 17, 39–51. Strande,N.T., Wyaj,D.W., Pryor,J.M. and Ramsden,D.A. (2014) Loparo,J.J. (2023) Holding it together: DNA end synapsis during non-homologous end joining. DNA Repair, 130, 103553. Watanabe,G. and Lieber,M.R. (2023) The flexible and iteragve steps within the NHEJ pathway. Biology, Biophysics 10.1016/j.pbiomolbio.2023.05.001. Molecular Progress and in Amin,H., Zahid,S., Hall,C. and Chaplin,A.K. (2024) Cold snapshots of DNA repair: Cryo-EM structures of DNA-PKcs and NHEJ machinery. Progress in Biophysics and Molecular Biology, 186, 1–13. Goff,N.J., Mikhova,M., Schmidt,J.C. and Meek,K. (2024) DNA-PK: A synopsis beyond synapsis. DNA Repair, 141, 103716. Rothkamm,K., Kruger,I., Thompson,L.H. and Lobrich,M. (2003) Pathways of DNA double- strand break repair during the mammalian cell cycle. Mol Cell Biol, 23, 5706–5715. Lees-Miller,J.P., Cobban,A., Katsonis,P., Bacolla,A., Tsutakawa,S.E., Hammel,M., Meek,K., Anderson,D.W., Lichtarge,O., Tainer,J.A., et al. (2021) Uncovering DNA-PKcs ancient phylogeny, unique sequence mogfs and insights for human disease. Progress in Biophysics and Molecular Biology, 163, 87–108. Sgnson,B.M., Moreno,A.T., Walter,J.C. and Loparo,J.J. (2020) A Mechanism to Minimize Errors during Non-homologous End Joining. Molecular Cell, 77, 1080-1091.e8. Deshpande,R.A., Myler,L.R., Soniat,M.M., Makharashvili,N., Lee,L., Lees-Miller,S.P., Finkelstein,I.J. and Paull,T.T. (2020) DNA-dependent protein kinase promotes DNA end processing by MRN and CtIP. Sci Adv, 6, 0922. Deshpande,R.A., Marin-Gonzalez,A., Barnes,H.K., Woolley,P.R., Ha,T. and Paull,T.T. (2023) Genome-wide analysis of DNA-PK-bound MRN cleavage products supports a sequengal model of DSB repair pathway choice. Nature communica9ons, 14. Chang,H. and Lieber,M. (2016) Structure-Specific nuclease acgviges of Artemis and the Artemis: DNA-PKcs complex. NUCLEIC ACIDS RESEARCH, 44, 4991–4997. Graham,T.G.W., Walter,J.C. and Loparo,J.J. (2016) Two-Stage Synapsis of DNA Ends during Non-homologous End Joining. Molecular Cell, 61, 850–858. Chen,S., Lee,L., Naila,T., Fishbain,S., Wang,A., Tomkinson,A.E., Lees-Miller,S.P. and He,Y. (2021) Structural basis of long-range to short-range synapgc transigon in NHEJ. Nature, 10.1038/s41586-021-03458-7. 78 14. 15. 16. 17. Chaplin,A.K., Hardwick,S.W., Liang,S., Kefala Stavridi,A., Hnizda,A., Cooper,L.R., De Oliveira,T.M., Chirgadze,D.Y. and Blundell,T.L. (2021) Dimers of DNA-PK create a stage for DNA double-strand break repair. Nat Struct Mol Biol, 28, 13–19. Seif-El-Dahan,M., Kefala-Stavridi,A., Frit,P., Hardwick,S.W., Chirgadze,D.Y., Maia De Oliviera,T., Brijon,S., Barboule,N., Bossaert,M., Pandurangan,A.P., et al. (2023) PAXX binding to the NHEJ machinery explains funcgonal redundancy with XLF. Science Advances, 9, eadg2834. Buehl,C.J., Goff,N.J., Hardwick,S.W., Gellert,M., Blundell,T.L., Yang,W., Chaplin,A.K. and Meek,K. (2023) Two disgnct long-range synapgc complexes promote different aspects of end processing prior to repair of DNA breaks by non-homologous end joining. Molecular Cell, 10.1016/j.molcel.2023.01.012. Goff,N.J., Brenière,M., Buehl,C.J., de Melo,A.J., Huskova,H., Ochi,T., Blundell,T.L., Mao,W., Yu,K., Modesg,M., et al. (2022) Catalygcally inacgve DNA ligase IV promotes DNA repair in living cells. Nucleic Acids Research, 10.1093/nar/gkac913. 18. Mikhova,M., Goff,N.J., Janovič,T., Heyza,J.R., Meek,K. and Schmidt,J.C. (2024) Single- molecule imaging reveals the kinegcs of non-homologous end-joining in living cells. 10.1101/2023.06.22.546088. 19. 20. 21. Sallmyr,A., Rashid,I., Bhandari,S.K., Naila,T. and Tomkinson,A.E. (2020) Human DNA ligases in replicagon and repair. DNA Repair, 93, 102908. Grawunder,U., Zimmer,D. and Lieber,M.R. (1998) DNA ligase IV binds to XRCC4 via a mogf located between rather than within its BRCT domains. Current Biology, 8, 873–879. Chaplin,A.K., Hardwick,S.W., Stavridi,A.K., Buehl,C.J., Goff,N.J., Ropars,V., Liang,S., Oliveira,T.M.D., Chirgadze,D.Y., Meek,K., et al. (2021) Cryo-EM of NHEJ supercomplexes provides insights into DNA repair. Molecular Cell, 0. 22. Sgnson,B.M., Carney,S.M., Walter,J.C. and Loparo,J.J. (2024) Structural role for DNA Ligase IV in promogng the fidelity of non-homologous end joining. Nat Commun, 15, 1250. 23. Malashejy,V., Au,A., Chavez,J., Hanna,M., Chu,J., Penna,J. and Cortes,P. (2023) The DNA binding domain and the C-terminal region of DNA Ligase IV specify its role in V(D)J recombinagon. PLOS ONE, 18, e0282236. 24. Medina-Suarez,D., Han,L., O’Reily,S., Liu,J., Wei,C., Brenière,M., Goff,N.J., Chen,C., Modesg,M., Meek,K., et al. (2024) DNA ligases 3 and 4 cooperate in vivo to facilitate DNA repair and organism viability. 25. Chan,S.H., Yu,A.M. and McVey,M. (2010) Dual Roles for DNA Polymerase Theta in Alternagve End-Joining Repair of Double-Strand Breaks in Drosophila. PLOS Gene9cs, 6, e1001005. 79 26. Wyaj,D.W., Feng,W., Conlin,M.P., Yousefzadeh,M.J., Roberts,S.A., Mieczkowski,P., Wood,R.D., Gupta,G.P. and Ramsden,D.A. (2016) Essengal Roles for Polymerase theta- Mediated End Joining in the Repair of Chromosome Breaks. Mol. Cell, 63, 662–673. 27. 28. 29. Yousefzadeh,M.J., Wyaj,D.W., Takata,K., Mu,Y., Hensley,S.C., Tomida,J., Bylund,G.O., Doublié,S., Johansson,E., Ramsden,D.A., et al. (2014) Mechanism of Suppression of Chromosomal Instability by DNA Polymerase POLQ. PLOS Gene9cs, 10, e1004654. Chen,S., Vogt,A., Lee,L., Naila,T., McKeown,R., Tomkinson,A.E., Lees-Miller,S.P. and He,Y. (2023) Cryo-EM visualizagon of DNA-PKcs structural intermediates in NHEJ. Science Advances, 9, eadg2838. Chen,X., Ballin,J.D., Della-Maria,J., Tsai,M.-S., White,E.J., Tomkinson,A.E. and Wilson,G.M. (2009) Disgnct kinegcs of human DNA ligases I, IIIα, IIIβ, and IV reveal direct DNA sensing ability and differengal physiological funcgons in DNA repair. DNA Repair, 8, 961–968. 30. Meek,K. (2020) Acgvagon of DNA-PK by hairpinned DNA ends reveals a stepwise mechanism of kinase acgvagon. Nucleic Acids Research, 48, 9098–9108. 31. Neal,J.A., Xu,Y., Abe,M., Hendrickson,E. and Meek,K. (2016) Restoragon of ATM Expression in DNA-PKcs–Deficient Cells Inhibits Signal End Joining. J.I., 196, 3032–3042. 32. Deriano,L., Stracker,T., Baker,A., Petrini,J. and Roth,D. (2009) Roles for NBS1 in Alternagve Nonhomologous End-Joining of V(D)J Recombinagon Intermediates. MOLECULAR CELL, 34, 13–25. 80 Chapter 4: Discussing the two-stage synapZc model of NHEJ. By Noah J. Goff Conclusions and Future Direcgons 81 INTRODUCTION My goal with this chapter is to place each of the previous chapters more firmly in the context of the evolving synapgc model of NHEJ, highlight some outstanding quesgons in the field, and to propose experiments involving DNA Ligase IV to address those quesgons. The overarching themes of my dissertagon research center around how DSB repair pathways—in pargcular end joining pathways—use structural features to promote repair while minimizing mutagons. Of course, we are not the only group acgvely working on this problem, and I’ve highlighted several notable studies released over the course of my PhD studies that contribute to our most recent model of NHEJ and how it interacts with alternagve end joining pathways. DISCUSSION Chapter 1: Evolving models of non-homologous end joining. The core of this chapter is a review of recent NHEJ literature that’s contributed to an updated model of end synapsis and processing. This work spans early single molecule imaging (SMI) studies performed by Loparo and colleagues (21, 46), early Cryo-EM work from Tom Blundell, Yuan He, and Amanda Chaplin (18, 19), and an introducgon to the role of DNA Ligase IV (L4). I’ve also reproduced secgons of a review that I co-wrote with another graduate student, Mariia Mikhova, and our advisors Kathy Meek and Jens Schmidt. The engre review is quite large (too large to reproduce in total for this work) and focuses on the role of DNA-PKcs in the new synapgc models of NHEJ. The secgons included were both primarily dramed by me and highly pergnent to understanding evolving models of NHEJ—a core throughline of both my dissertagon work and other contribugons during my PhD. There have been several other reviews from leading groups focused on other aspects of recent end joining research—including excellent reviews of recent Cryo-EM data (52, 53), and end-synapsis (80). Chapter 2: CatalyZcally inacZve DNA ligase IV promotes DNA repair in living cells. This project began as a straighiorward CRISPR-Cas9 gene edigng experiment coupled with an easy set of lab-standard reporter assays to onboard a first-year graduate student. We never expected to be the core project of my PhD. The inigal 293T mutagenesis work in early spring of 2020, immediately before the onset of the COVID-19 pandemic. The central research quesgon—does catalygcally inacgve L4 promote end joining in cell cultures—was inspired by papers from 82 Loparo and colleagues proposing a 2-stage synapgc model of NHEJ, where DNA ends were “protected” in a long range complex prior to recruitment of downstream factors (21, 46). Importantly, the authors discovered that L4’s catalygc funcgon was dispensable for this result in vitro, indicagng L4 held important noncatalygc funcgon that could support otherwise highly regulated end processing. While other studies had suggested a non-catalygc role for L4 both in vitro and in yeast cells, it was unclear whether DSB repair in more complex eukaryogc cells would support any NHEJ amer loss of L4’s core catalygc funcgon. Furthermore, the studies only tested the role of L4 in promogng processing mediated by DNA polymerase lambda and the phosphodiesterase TDP1. In addigon to studying NHEJ, our lab also has research projects into immunoglobulin development through VDJ recombinagon, which produces NHEJ-targeted DNA hairpin structures as recombinagon intermediates. Given the addigonal targegng of hairpins to NHEJ, we were quite surprised when the inigal 293T joining assays revealed robust hairpin joining in cells lacking L4 catalygc acgvity. Notably, the inigal in vitro work from Loparo and colleagues did not test DNA hairpin substrates in the biochemical xenopus with catalygcally inacgve L4. While revising the paper, we found a 2013 paper from Wilson and colleagues (90) that provided early evidence for catalygcally inacgve L4 supporgng end joining in yeast. We were completely unaware of this paper at the gme, but it suggests a remarkable conservagon of NHEJ phenotypes across eukaryotes—even those lacking DNA-PKcs. The primary takeaways from my first paper were: 1) that L4’s structural presence does serve as a regulatory step in canonical NHEJ, 2) loss of L4 catalygc acgvity in the shortrange complex decreases the fidelity of end joining, and 3) that the L3-XRCC1 complex acts collaboragvely with the core NHEJ factors in vitro to sgmulate end-joining. To date, the role of a L3-NHEJ connecgon serves as a major extant quesgon in the two-stage iteragve model of NHEJ. An in-review (at gme of wrigng) paper from Dr. Kefei Yu’s laboratory established a catalygcally inacgve L4K273S/K273S and L3nuc-/- mouse lines, with each being viable through birth and adolescence. Notably, L3nuc-/- L4K273S/K273S double mutant offspring are nonviable, providing addigonal in vivo evidence that there is compensagon from L3 following loss of L4’s catalygc acgvity. 83 Chapter 3: New insight into how the DNA binding domain of DNA Ligase IV facilitates end joining independent of its catalyZc acZvity. This project originated both as a direct follow-up to my first publicagon, but also a structure-directed invesggagon of new data from Amanda Chaplin and Steve Hardwick’s group at the University of Leicester and Cambridge University to invesggate interfaces discovered in new Cryo-EM data. It is my current area of acgve research. We have shown that disrupgng end-tethering substangally impairs L4 K273A-mediated end joining, with only modest impacts on joining in the presence of catalygcally acgve L4. I propose that tethering ends in the SRC occurs through four mechanisms: 1) the XRCC4:XLF:XRCC4 bridge linking each Ku70/80 heterodimer, 2) the trans L4 DBD:Ku70 interacgon (disrupted by the RK>DD mutagon) that anchors one end of the complex with the ligagon posigoned break, 3) the L4-DNA interacgon recently reported by Loparo and colleagues (48), and 4) rapid ligagon of one strand early following transigon into the shortrange complex to covalently link DNA ends. As seen in chapter 3, disrupgon of any two of these interfaces appears to be sufficient to ablate end joining, either mediated by L4 catalygc acgvity or through promogon of alt-EJ pathways (as is the case with catalygcally inacgve L4). In addigon to invesggagng interfaces that could mediate L4’s structural role in NHEJ, we also sought to invesggate backup repair factors (in addigon to L3-mediated joining) that promote end joining amer failed NHEJ. Based on my observagon that cells expressing catalygcally inacgve L4 overuglizes microhomology in L4 K273A cells (Chapter 2), we hypothesized that DNA- Polymerase Theta (Polθ) may play an outsized role in supporgng NHEJ in cases where the ligagon complex fails. To test, I used a CRISPR/Cas9 genome edigng approach to knock out Polθ from my 293T L4K273A*/- cells, then tested their response to a MMEJ specific reporter substrate, NHEJ- specific VDJ Coding joint substrate, and sensigvity to DSB inducing calicheamicin and teniposide. Of note, I was unable to isolate any 293T L4-/- Polθ-/- double knockout cell strains despite mulgple gRNA transfecgons. Based on previous experiments in the lab as well as other reports (77, 91, 92), we believe that Polθ serves as the primary alternagve end joining factor following loss of NHEJ and as such double knockouts are nonviable—or nearly nonviable. This hypothesis is supported by my experiments indicagng that 293T L4K273A/- Polθ-/- cells are hypersensigve to DSB inducing agents calicheamicin and teniposide (Chapter 3). Anecdotally, 84 these cell cultures also divide much slower, comparable to L4-/- cultured under the same condigons. There is neither a growth disparity nor DSB drug sensigvity observed with loss of Polθ on its own. Despite the hypersensigvity to genomic DSBs, addigonal loss of Polθ appears to have no impact on VDJ coding, and only a modest deficiency in joining TelN and I-SceI substrates. A combinagon of two factors could explain this: 1) signal from episomal joining assays is measured as a binary on/off per cell and is not necessarily a fair measure of total break repair capacity for a given substrate so long as minimal joining is performed; 2) there may be an alternate repair mechanism that can join simple overhangs (like those generated by I-SceI and Artemis-opened hairpins) but not more complex adducts caused by calicheamicin and teniposide. I suggest that L3 may serve in this role based on Chapter 2 and the mouse models developed by Dr. Kefei Yu’s laboratory. AddiZonal contribuZons to work that haven’t been reproduced for my dissertaZon: In addigon to the work presented in chapters 2 and 3, I have made several substangal contribugons to other projects that have resulted in authorship, both published and in press. I’ve chosen to focus my dissertagon on my Ligase IV projects, and as such I’ve not reproduced excerpts from these manuscripts (a full list of my contribugons to papers can be found in Appendix 1). Ku80 Linker Mutants: First, I’d like to highlight my contribugons to tesgng the Ku80 C-terminal domain mutagons published in Buehl et al. 2023 (20). This project began shortly amer returning from COVID-19 lockdowns, with our lab working collaboragvely with several structural biologists to use mutagonal strategies to test the funcgonal relevance of synapgc NHEJ complex structures recently idengfied using Cryo-EM. These structures revealed a trans interacgon between Ku80’s acidic extreme C-terminal domain (CTD) and a basic patch on DNA-PKcs, mediated by an unstructured flexible linker in Ku80 that is highly conserved in length (but not primary sequence). The Ku80 CTD had previously been idengfied as a mediator of end joining (5–7), and mutagng the DNA-PKcs residues that mediate this interacgon (termed 4A) revealed a significant defect in nucleolygc end processing. We hypothesized that the highly conserved length of the flexible linker (93) observed in the Cryo-EM structures may be funcgonally relevant in promogng the trans interacgon. It was observed by Dr. Wei Yang (NIH) that the Ku80 CTD could also interact with DNA-PKcs in cis if the linker stretches to its maximum length, and 85 she suggested that adding or removing residues may differengally impact funcgon by biasing formagon of the cis or trans interacgons. We observed that lengthening the linker by 6-15 residues substangally reduced the amount of joining (>2 fold). Interesgngly, shortening the linker 3 or 6 residues significantly increased NHEJ funcgon both in episomal joining assays and in resistance to calicheamicin and etoposide (a Top2 poison). Further shortening the linker by 10, 13, or 15 residues progressively impaired NHEJ, with the shortest linker length mimicking complete delegon of the CTD. We concluded that the hypothesized cis interacgon may have an inhibitory effect on synapsis, and there is a minimum linker length required to promote synapsis and NHEJ funcgon. It’s currently unclear why the flexible linker is conserved at its current length (as opposed to 3-6 AA shorter), it may be due to lower repair fidelity, or some other deficiency not idengfied in our tesgng. Single Molecule Imaging of NHEJ Factors: The second contribugon I would like to highlight is to Mariia Mikhova’s fantasgc study into the nuclear dynamics of NHEJ factors using live-cell Single Molecule Imaging (73), in pargcular my work generagng L4 knockouts in their U2OS cell strains with halo-tagged NHEJ factors. These cells were then used to assess changes in Ku70, DNA-PKcs, XLF, and XRCC4 dynamics with complementagon of L4 expression vectors for the revisions of her (in press) paper. Notable findings include: 1) XRCC4 is primarily localized to the nucleus but does not interact with chromagn in the absence of L4, 2) There appears to be a mechanism to acgvely remove Ku and DNA-PKcs if L4/XRCC4 recruitment fails, and 3) in the presence of catalygcally inacgve L4 all four tagged NHEJ factors have a greatly elongated window (compared to WT L4) where they interact with chromagn—including DNA-PKcs. This final point is pargcularly surprising given that DNA-PKcs departs chromagn significantly earlier than other factors. We originally hypothesized that DNA-PKcs departure corresponded with transigon into the short-range complex—matching the single Cryo-EM structure reported by Yuan He and colleagues (18), but the in vitro work indicates that there is no defect in short-range complex formagon or in end processing in the presence of catalygcally inacgve L4 (21, 46). In my opinion, it’s likely that the Cryo-EM short-range complex structure is not necessarily the only possible conformagon of NHEJ factors in the experimentally observed short-range complex. To wit, I believe that it may be possible for DNA-PKcs to remain present in some short-range 86 complexes, but it notably does depart the break earlier than other factors— possibly amer single strand ligagon to allow more room for processing the second strand. Ongoing argument over the role of DNA-PKcs and an alternate model of synapsis: DNA-PKcs had previously thought to primarily be dominant in NHEJ in vertebrates— pargcularly in those that predominantly use VDJ recombinagon for immunoglobulin diversificagon. Central to this hypothesis was the absence of DNA-PKcs from several model organisms: most notably budding yeast (S. cerevisiae), C. elegans, and fruit flies (D. melanogaster). A study from Lees-Miller and colleagues searched genomic sequencing data across a wide range of species for potengal DNA- PKcs orthologs looking to build a deeper understanding of how NHEJ evolved (35). Remarkably, the authors discovered candidate DNA-PKcs orthologs in genomes sequenced from a broad range of plant, fungi, and animal species—with high conservagon of some well-studied features (including the ABCDE phosphorylagon patch). This startling result suggests that DNA-PKcs is not a unique regulator of NHEJ that arose due to programmed recombinagon—but rather a more universal regulator of core end joining across nucleated life-forms. There is an alternate model for NHEJ synapsis and ligagon—primarily championed by Eli Rothenberg (NYU) and Michael Lieber (USC). They conclude—through their imaging approaches using purified NHEJ proteins—that DNA-PKcs is dispensable for most NHEJ funcgon and tethering may occur through XRCC4-XLF filaments coagng intact secgons of DNA (they observe filaments of purified LX4 complexes coagng dsDNA near a DNA end with super resolugon microscopy). In their model, Ku70/80 recognizes one end, promotes filamentagon upstream of the break, then Ku binds to a trans filament (bound to the opposite DNA end) to inigate synapsis, then proceeds to bring the ends into a short-range synapse (measured as direct FRET+ signal in their assays). Notably, addigon of DNA-PKcs reduces the number of FRET+ foci (decreasing their “pairing efficiency” quangficagon) but does substangally increase the size of each focus. The authors claim that the presence of DNA-PKcs only results in pairing of “clusters” of DSBs, and that their relagvely low measurement of “normalized pairing efficiency” points to DNA-PKcs being dispensable for “simple” DSBs (60). I propose an alternate hypothesis where DNA-PKcs can act to synapse the DNA-stem loop structures used to cap their fluorescent oligonucleogdes (the authors confirmed that purified Ku70/80 + LX4 was unable to synapse these hairpin loops, but never ran 87 the control in the presence of DNA-PKcs). The stem-loop structures closely resemble hairpin DNA intermediates present in VDJ recombinagon, and the resulgng aggregate of tagged DNA donors would perfectly match the larger foci seen in all reacgons included with DNA-PKcs. Furthermore, data from Mariia Mikhova’s paper indicate that there is likely no role for XRCC4-XLF filaments given XRCC4 does not associate with breaks in the absence of L4 (73). In my opinion, most of their reasoning regarding the role of DNA-PKcs is circular; the authors cite the data from Reid et al. 2015 (60) as jusgficagon to not test if DNA-PKcs promotes synapsis in subsequent papers (42, 58, 59). They then use their engre body of work to argue against DNA-PKcs’s involvement in most NHEJ events. Their experimental design is unable to independently visualize localizagon of the acceptor fluorophore in the absence of a donor in proximity for FRET (less than 100 Å with their fluorophores), therefore making it impossible to clearly observe long-range synapses. This approach is less powerful than the Loparo group’s studies and misses synapsis consistent with the LRC (where ends are not held close enough for the FRET+ signal Rothenberg and colleagues uglized as their metric for synapsis). In their measurements, they observe that addigon of purified Ku70/80, XLF, and L4-XRCC4 is sufficient to synapse ends using a “Normalized Frequency” of high-FRET foci. The authors argue for a model of NHEJ resolving “simple DSBs” without presence or acgvity of DNA-PKcs. Instead, they argue synapsis occurs through long XLF-XRCC4-L4 filaments that they observe “coagng” DNA. Studies on the Zming of NHEJ are a major component of the new two-stage synapZc model: A recent report from Loparo and colleagues addressed the dynamics of L4 at the instant of transigon into the short-range complex. They developed a three-color imaging system to simultaneously DNA end synapsis (consistent with the SRC) as well as DNA-protein interacgons. Their data suggest that two molecules of L4 are recruited to a DSB prior to complex formagon and both dynamically interact with DNA ends. Prior to short-range synapsis, one LX4 complex vacates the LRC seconds before they observe DNA-DNA interacgons consistent with SRC formagon (48). Unfortunately, the authors only tested substrates generated with blunt ligagon- compagble DNA ends that require no addigonal end-processing. I believe it’s likely that one molecule of L4 may act to catalyze single strand break repair—pargcularly given that L4 is pre- adenylated in a catalygcally charged state it seems likely that this reacgon may progress very 88 quickly. Tesgng a wider range of end joining substrates in the three-color smFRET experiment— pargcularly a substrate that requires short-range complex targeted end processing—would be highly informagve. Addigonal open quesgons remain regarding the role of DNA-PKcs in supporgng the short- range complex. The inigal report of a short-range complex observed without DNA-PKcs by Cryo- EM (18), coupled with preliminary data showing DNA-PKcs leaving chromagn earlier than other NHEJ factors in a SMI study (16) suggested to us that DNA-PKcs dissociated from the break site amer inigal synapsis and end processing. One striking result: DNA-PKcs remains bound to breaks for significantly longer with catalygcally inacgve L4. It’s unclear whether DNA-PKcs’s retengon at breaks is mediated through a prolonged long range complex (something unsupported by the in vitro smFRET data) (21, 46) or if there is a conformagon of the short-range complex that retains DNA-PKcs. I suggest the following model to explain the discrepancy in data: DNA-PKcs remains to stabilize the short-range complex ungl L4 can catalyze repair of one end. Finalizing catalysis allows for L4 and DNA-PKcs to dissociate from the break, leaving behind a significantly less hazardous break that can be repaired by the remaining NHEJ machinery (XRCC4, XLF, and Ku all remain bound for significantly longer than DNA-PKcs). This turnover of DNA-PKcs would allow it to “supervise” repair, then either recycle to bind another DSB or allow more room for extensive end processing on the remaining SSB (pargcularly in a case following ligagon opposite another DNA lesion: gap, mismatch, aberrant base, etc.). 89 Figure 19: NHEJ progresses through a two-stage synapZc mechanism. Normal NHEJ progresses from break recognigon (DNA-PK monomers) to LRC formagon. Depending on end chemistry, either the end-protecgon XLF mediated dimer (for ends with small adducts/overhangs ends, middle lem) or the Ku80-mediated dimer (ends with bulky adducts, middle right). The NHEJ synapse transigons into a SRC following recruitment of LX4 and XLF (and possibly formagon of the XLF-mediated dimer). Loss of L4 catalygc acgvity promotes long-term stable SRC synapsis, leading to excessive end processing followed by Polθ/L3 mediated end joining (bojom right). 90 Figure 19 (cont’d) Notably, loss of tethering acgvity (either of the mDBD or RK>DD mutagons discussed in chapter 3) in the SRC in combinagon with losing L4 catalygc acgvity results in no L4-mediated alt-EJ. Furthermore, catalygcally acgve L4 combining DNA end binding mutagons (mDBD) with the L4- Ku70 interface mutant (RK>DD) ablates nearly all NHEJ. Areas of future L4 research: While I believe the work in this dissertagon outlines an important contribugon to our understanding of NHEJ, there are several open quesgons regarding L4’s role in end joining that I would like to see addressed in the future. First, addigonal work uglizing many of the newly idengfied mutants in Chapter 3 could explain why targegng the shared interfaces impact NHEJ so severely. I propose performing a suite of single-molecule imaging studies— idengcal to the calicheamicin “gme course” studies using transiently complemented L4 that I contributed to in Mariia Mikhova’s study (73)—pargcularly to study nuclear dynamics of XRCC4 and DNA-PKcs in the short-range complex. I hypothesize that disrupgng residues important for stable long-term synapsis should shorten the gme that the XRCC4-L4 complex remains bound to breaks, resulgng in a shortened window for chromagn associagon. Conversely, the gme-window for DNA-PKcs chromagn associagon should increase as failed repair events require rebinding and synapsis before beginning the pathway again. To support this model, I suggest that future researchers should perform deep sequencing across repaired breaks to invesggate repair outcomes in a disrupted shortrange complex. Our current model of NHEJ predicts that the high fidelity of joining rougne breaks can be ajributed to highly regulated access to DNA ends prior to ligagon. It would be very interesgng to know if a disrupgon in short-range complex stability (which has a modest impact on joining of most breaks, chapter 3) also disrupts fidelity of repair outcomes. Furthermore, combining L4 K273A with the RK>DD mutant (disrupgng the L4-Ku70 interacgon in the SRC) ablates nearly all joining. In my 2022 paper, we suggested that prolonged synapsis in the short-range complex could contribute to excessive end processing and the increased rate of delegons. Perhaps the repair outcomes in these combinagon mutants are both less efficient at repairing breaks but higher fidelity than the forms of alternagve end joining. I also suggest that a future graduate student uglize the Ku70/DNA-PK/XLF/XRCC4 halo + L4 to test mutagons idengfied by other groups. The SMI approach in these cells allows 91 conclusions regarding the gming and regulagon of NHEJ that are currently unaddressed. The recent L4 DBD-DNA interacgng mutants reported in Sgnson et al. 2024 (48) clearly disrupt short- range complex formagon in vitro, but there are important differences between biochemical systems and live-cell imaging. For example, deplegon of XRCC4-L4 complex (LX4) from frog extracts has minor impacts in the frog extract system has no discernable impact on LR complex formagon (46); however, there are clear major impacts on DNA-PKcs and Ku70’s associagon with chromagn in live cells, hypothesized to be dissolugon of the LRC if LX4 is unable to associate with breaks (73). To this end, I propose uglizing the DNA-PKcs-, Ku70-, and XRCC4-halo L4-/- cell strains to test the L4 DBD-DNA interacgng mutants published in S9nson et al. 2024 (48). I predict that targegng the proposed L4-DNA interacgon may trap NHEJ complexes in the LR complex in a manner that protects from NHEJ-aborgon, resulgng in prolonged associagon of all three tagged NHEJ factors. Beyond imaging canonical NHEJ factors in the context of L4 mutants, it also would be interesgng to image alternagve end joining factors both 1) in the presence of catalygcally inacgve L4 to assess gming of alt-EJ amer failed NHEJ and 2) in the presence of fully funcgonal L4 to determine whether tradigonal “backup” factors respond to DSB inducgon. There is an open quesgon from chapters 2 and 3 whether XRCC1-L3 can respond to DSBs and act directly within the short-range complex in vivo. I briefly ajempted a version of this experiment by transiently transfecgng L4-/- U2OS cells with expression vectors containing L4 K273A and L3-Halo (data not shown). Unfortunately, there were inconsistencies with L3-Halo expression levels, and I was unable to perform any experiments to test response to calicheamicin. A more careful experimental setup—likely generagng a genomically edited halo-L3 L4-/- cell strain may be required to fully address this quesgon. Beyond imaging approaches, I think there is significant work to be done understanding both what processes promotes NHEJ’s high fidelity outcomes and what factors determine pathway choice between NHEJ and mutagenic alternagve end joining. Experiments with a substrate that assesses joining via 7 bp of microhomology (Chapter 3) suggest that a certain subset of DSBs are joined via a highly mutagenic Polθ-dependent mechanism—even in the presence of NHEJ. Using high-depth Illumina sequencing, both my work and work from other labs 92 (21, 20, 94, 10) indicates that NHEJ+ cells generally repair breaks with extremely high fidelity—or at least consistently. Further experiments looking at mutagonal frequency from resolved chromosomal and episomal breaks in the Polθ +/- cells, pargcularly the L4K273A*/- cells to examine the quality of repair outcome following break inducgon. Preliminary sequencing results are inconclusive, VDJ coding substrates were a poor choice. While several papers have addressed the fidelity of NHEJ in small scale sequencing studies, I believe decreasing sequencing costs will enable larger whole-genome sequencing projects to invesggate how mutagons in NHEJ impact repair fidelity genome-wide. To do this, I propose sequencing whole-genomes of cells treated +/- DSB inducing agents in NHEJ mutant cell lines. This could become a valuable complementary approach to smaller targeted experiments like the presented targeted sequencing and would deepen our understanding of how mutagons in NHEJ factors impact genomic stability—pargcularly those which have been idengfied as funcgonally significant in lab or prevalent in the clinic. Addigonally, newer sequencing techniques— pargcularly Oxford Nanopore Technologies can inform not only per-base data but also provide more accurate representagons of genomic translocagons and copy number variagons for repeggve regions. FINAL THOUGHTS First and foremost, thank you for reading through this dissertagon, I hope you appreciate both the amount of work that’s been done across the field over the past five years and my contribugons to understanding L4’s role in the evolving NHEJ model. Graduate school during the COVID pandemic was not easy, between lockdowns limigng access to the lab (a significant hurdle when first isolagng and screening clones from my inigal L4 CRISPR experiment) to supply shortages delaying experiments. Over the course of my PhD, I was most surprised by the literature revealing how prevalent DNA-PKcs across eukaryotes. When I first met Kathy in early September 2019 (before nearly all of the most recent studies into NHEJ’s synapgc mechanism), I recall talking about how the mysteries of DNA-PKcs expression levels in humans compared to its absence in many other well studied organisms. This lem major doubts about how widely we could apply our studies beyond vertebrates. Learning the true breadth of cross species DNA-PKcs conservagon— from plants to fungi to insects and beyond—points to some deeper throughline of highly 93 regulated end joining repair and deepens my appreciagon for NHEJ’s role across eukaryotes. To that end, I hope that research into DNA Ligase IV’s role in promogng end-joining congnues in groups at MSU and beyond. 94 REFERENCES 1. 2. 3. 4. 5. 6. 7. 8. 9. Graham,T.G.W., Walter,J.C. and Loparo,J.J. (2016) Two-Stage Synapsis of DNA Ends during Non-homologous End Joining. Molecular Cell, 61, 850–858. Sgnson,B.M., Moreno,A.T., Walter,J.C. and Loparo,J.J. (2020) A Mechanism to Minimize Errors during Non-homologous End Joining. Molecular Cell, 77, 1080-1091.e8. Chen,S., Lee,L., Naila,T., Fishbain,S., Wang,A., Tomkinson,A.E., Lees-Miller,S.P. and He,Y. (2021) Structural basis of long-range to short-range synapgc transigon in NHEJ. Nature, 10.1038/s41586-021-03458-7. Chaplin,A.K., Hardwick,S.W., Stavridi,A.K., Buehl,C.J., Goff,N.J., Ropars,V., Liang,S., Oliveira,T.M.D., Chirgadze,D.Y., Meek,K., et al. (2021) Cryo-EM of NHEJ supercomplexes provides insights into DNA repair. Molecular Cell, 0. Amin,H., Zahid,S., Hall,C. and Chaplin,A.K. (2024) Cold snapshots of DNA repair: Cryo-EM structures of DNA-PKcs and NHEJ machinery. Progress in Biophysics and Molecular Biology, 186, 1–13. Vogt,A., He,Y. and Lees-Miller,S.P. (2023) How to fix DNA breaks: new insights into the mechanism of non-homologous end joining. Biochemical Society Transac9ons, 10.1042/BST20220741. Loparo,J.J. (2023) Holding it together: DNA end synapsis during non-homologous end joining. DNA Repair, 130, 103553. Chiruvella,K.K., Liang,Z., Birkeland,S.R., Basrur,V. and Wilson,T.E. (2013) Saccharomyces cerevisiae DNA Ligase IV Supports Imprecise End Joining Independently of Its Catalygc Acgvity. PLOS Gene9cs, 9, e1003599. Sgnson,B.M., Carney,S.M., Walter,J.C. and Loparo,J.J. (2024) Structural role for DNA Ligase IV in promogng the fidelity of non-homologous end joining. Nat Commun, 15, 1250. 10. Wyaj,D.W., Feng,W., Conlin,M.P., Yousefzadeh,M.J., Roberts,S.A., Mieczkowski,P., Wood,R.D., Gupta,G.P. and Ramsden,D.A. (2016) Essengal Roles for Polymerase theta- Mediated End Joining in the Repair of Chromosome Breaks. Mol. Cell, 63, 662–673. 11. Merker,L., Feller,L., Dorn,A. and Puchta,H. Deficiency of both classical and alternagve end- joining pathways leads to a synergisgc defect in double-strand break repair but not to an increase in homology-dependent gene targegng in Arabidopsis. The Plant Journal, n/a. 12. 13. Zhou,J., Gelot,C., Pantelidou,C., Li,A., Yücel,H., Davis,R.E., Färkkilä,A., Kochupurakkal,B., Syed,A., Shapiro,G.I., et al. (2021) A first-in-class polymerase theta inhibitor selecgvely targets homologous-recombinagon-deficient tumors. Nat Cancer, 2, 598–610. Buehl,C.J., Goff,N.J., Hardwick,S.W., Gellert,M., Blundell,T.L., Yang,W., Chaplin,A.K. and Meek,K. (2023) Two disgnct long-range synapgc complexes promote different aspects of end processing prior to repair of DNA breaks by non-homologous end joining. Molecular Cell, 10.1016/j.molcel.2023.01.012. 95 14. 15. Inagawa,T., Wennink,T., Lebbink,J., Keijzers,G., Florea,B., Verkaik,N. and van Gent,D. (2020) C-Terminal Extensions of Ku70 and Ku80 Differengally Influence DNA End Binding Properges. INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, 21. Singleton,B.K., Torres-Arzayus,M.I., Ro~nghaus,S.T., Taccioli,G.E. and Jeggo,P.A. (1999) The C Terminus of Ku80 Acgvates the DNA-Dependent Protein Kinase Catalygc Subunit. Mol. Cell. Biol., 19, 3267–3277. 16. Weterings,E., Verkaik,N.S., Keijzers,G., Florea,B.I., Wang,S.-Y., Ortega,L.G., Uematsu,N., Chen,D.J. and van Gent,D.C. (2009) The Ku80 Carboxy Terminus Sgmulates Joining and Artemis-Mediated Processing of DNA Ends. MCB, 29, 1134–1142. 17. Chen,S., Lees-Miller,J.P., He,Y. and Lees-Miller,S.P. (2021) Structural insights into the role of DNA-PK as a master regulator in NHEJ. GENOME INSTAB. DIS., 2, 195–210. 18. Mikhova,M., Goff,N.J., Janovič,T., Heyza,J.R., Meek,K. and Schmidt,J.C. (2024) Single- molecule imaging reveals the kinegcs of non-homologous end-joining in living cells. 10.1101/2023.06.22.546088. 19. 20. 21. 22. 23. 24. 25. Lees-Miller,J.P., Cobban,A., Katsonis,P., Bacolla,A., Tsutakawa,S.E., Hammel,M., Meek,K., Anderson,D.W., Lichtarge,O., Tainer,J.A., et al. (2021) Uncovering DNA-PKcs ancient phylogeny, unique sequence mogfs and insights for human disease. Progress in Biophysics and Molecular Biology, 163, 87–108. Reid,D.A., Keegan,S., Leo-Macias,A., Watanabe,G., Strande,N.T., Chang,H.H., Oksuz,B.A., Fenyo,D., Lieber,M.R., Ramsden,D.A., et al. (2015) Organizagon and dynamics of the nonhomologous end-joining machinery during DNA double-strand break repair. Proceedings of the Na9onal Academy of Sciences, 112, E2575–E2584. Reid,D.A., Conlin,M.P., Yin,Y., Chang,H.H., Watanabe,G., Lieber,M.R., Ramsden,D.A. and Rothenberg,E. (2017) Bridging of double-stranded breaks by the nonhomologous end- joining ligagon complex is modulated by DNA end chemistry. Nucleic Acids Research, 45, 1872–1878. Conlin,M.P., Reid,D.A., Small,G.W., Chang,H.H., Watanabe,G., Lieber,M.R., Ramsden,D.A. IV Guides End-Processing Choice during and Rothenberg,E. (2017) DNA Ligase Nonhomologous End Joining. Cell Reports, 20, 2810–2819. Zhao,B., Watanabe,G., Morten,M.J., Reid,D.A., Rothenberg,E. and Lieber,M.R. (2019) The essengal elements for the noncovalent associagon of two DNA ends during NHEJ synapsis. Nat Commun, 10, 3588. Luedeman,M.E., Stroik,S., Feng,W., Luthman,A.J., Gupta,G.P. and Ramsden,D.A. (2022) Poly(ADP) ribose polymerase promotes DNA polymerase theta-mediated end joining by acgvagon of end resecgon. Nat Commun, 13, 4547. Goff,N.J., Brenière,M., Buehl,C.J., de Melo,A.J., Huskova,H., Ochi,T., Blundell,T.L., Mao,W., Yu,K., Modesg,M., et al. (2022) Catalygcally inacgve DNA ligase IV promotes DNA repair in living cells. Nucleic Acids Research, 10.1093/nar/gkac913. 96 APPENDIX: A list of authorship contribugons to other papers. This is a list of authorship contribugons to academic papers as described in Chapter 4. All informagon updated as of 11-25-2024, presented in reverse chronological order of acceptance for publicagon. 1. Medina-Suarez, D., Han, L., O’Reily, S., Liu, J., Wei, C., Brenière, M., Goff, N.J., Chen, C., Modesg, M., Meek, K., Harrington, B., Yu, K. (2024). DNA ligases 3 and 4 cooperate in vivo to facilitate DNA repair and organism viability. Nucleic Acids Research, In Press. 2. Mikhova, M., Goff, N.J., Janovič, T., Heyza, J.R., Meek, K., Schmidt, J.C. (2024) Single- molecule imaging reveals the kinegcs of non-homologous end-joining in living cells. Nature Communica9ons, in press. 3. Pascarella, G., Conner, K.N., Goff, N.J., Carninci,P., Olive,A.J. & Meek,K. (2024) Compared to other NHEJ factors, DNA-PK protein and RNA levels are markedly increased in all higher primates, but not in prosimians or other mammals. DNA Repair, 142, 103737. 4. Goff, N.J., Mikhova, M., Schmidt, J.C., & Meek, K. (2024). DNA-PK: A synopsis beyond Synapsis. DNA Repair, 141, 103716. 5. Buehl, C. J., Goff, N.J., Hardwick, S.W., Gellert, M., Blundell, T.L., Yang, W., Chaplin, A.K., & Meek, K. (2023). Two disgnct long-range synapgc complexes promote different aspects of end-processing prior to repair of DNA breaks by non-homologous end joining. Molecular Cell 83(5), 698-714. 6. Goff, N. J., Brenière, M., Buehl, C. J., de Melo, A. J., Huskova, H., Ochi, T., Blundell, T, Weifeng, M., Yu, K., Modesg, M., & Meek, K. (2022) Catalygcally inacgve DNA ligase IV promotes DNA repair in living cells. Nucleic Acids Research, 50(19), 11058–11071. 7. Liu, L., Chen, X., Li, J., Wang, H., Buehl, C. J., Goff, N. J., Meek, K., Yang, W., & Gellert, M. (2022). Autophosphorylagon transforms DNA-PK from protecgng to processing DNA ends. Molecular Cell, 82(1), 177-189. 8. Chaplin, A. K., Hardwick, S. W., Stavridi, A. K., Buehl, C. J., Goff, N. J., Ropars, V., Liang, S., De Oliveira, T. M., Chirgadze, D. Y., Meek, K., Charbonnier, J.B., & Blundell, T. L. (2021). Cryo-EM of NHEJ supercomplexes provides insights into DNA repair. Molecular Cell, 81(16), 3400-3409. 97