Abstract
The DDR consists of multiple pathways that sense, signal, and respond to anomalous DNA. To promote efficient replication, viruses have evolved to engage and even modulate the DDR. In this review, we will discuss a select set of diverse viruses and the range of mechanisms they evolved to interact with the DDR and some of the subsequent cellular consequences. There is a dichotomy in that the DDR can be both beneficial for viruses yet antiviral. We will also review the connection between the DDR and innate immunity. Previously believed to be disparate cellular functions, more recent research is emerging that links these processes. Furthermore, we will discuss some discrepancies in the literature that we propose can be remedied by utilizing more consistent DDR-focused assays. By doing so, we hope to obtain a much clearer understanding of how broadly these mechanisms and phenotypes are conserved among all viruses. This is crucial for human health since understanding how viruses manipulate the DDR presents an important and tractable target for antiviral therapies.
Keywords: DNA damage response, innate immunity, viral-host interactions, viral replication, antiviral response
Introduction
To maintain genomic integrity, cells possess various mechanisms to repair, protect, and replicate genetic material. At the heart of this is the DNA damage response (DDR), a signaling cascade that functions to sense, signal, and respond to aberrant nucleic acid. However, in addition to maintaining genomic integrity, the DDR is exquisitely poised to regulate viral infection, since to the cell, viruses are essentially aberrant nucleic acids. In support of this, the connection between viral replication and the DDR has emerged in two primary roles: (1) viruses modulate the DDR proteins and pathways required for viral replication; (2) there is significant crosstalk between the DDR and the innate immune response against viruses. These connections have been observed extensively across viral classifications and are relevant to a variety of both DNA and RNA viruses, including single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA) viruses, positive (+) and negative (−) stranded RNA viruses, and retroviruses (a (+) RNA virus that relies on a dsDNA intermediate), which we separately classify here according to the Baltimore Classification.1
In this review, we will break down the interaction of viruses with the DDR into four primary sections (Figure 1). First, we will discuss how viruses induce DNA damage and antagonize the sensing of this DNA damage. Second, we will discuss how viruses modulate the DDR signaling cascade – from mediators, to transducers, to effectors – with a focus on specific DNA, RNA, and retroviruses. Third, we will highlight the many cellular consequences of viral-induced modulation of the DDR. Finally, we will connect DDR signaling to innate immunity to emphasize how they are two sides of the same coin, since both are signaling cascades that sense, signal, and respond to aberrant nucleic acids. Each “side of the coin” (cellular function) has been traditionally thought to be separate, but more recent work has shown them to be more interconnected. We will not be able to cite all the work that has gone into understanding the connections between viral replication and the DDR (for additional specialized topic reviews, see 2,3 and others highlighted throughout the text). Additionally, some of the data we cite is limited and has yet to be corroborated. We understand the limitations this brings, yet we have included the work to demonstrate that examples exist across diverse viruses to substantiate the larger themes and concepts we discuss. We aim for this review to serve not only as an additional resource but to stimulate the fields of virology, DDR, and innate immunity to look for potential crosstalk in these interconnected fields that are essential for human health.
Figure 1. Four major features of interplay between viruses and the DDR.

Viral engagement of the DDR embodies four major characteristics: Recognition, Signaling, Cellular Consequences, and Innate Immunity. Each of these four pillars can be further broken down into specific components that make up the larger characteristics that viruses have evolved to modulate either through a precise mechanism or as a consequence of infection. (1) Recognition initiates the DDR and is activated by DNA damage. Sensors recognize this DNA damage to activate ATM, ATR, or DNA-PK signaling. Viruses have evolved to modulate this step by inducing DNA damage and antagonizing damage sensors. (2) Signaling then occurs through a variety of mediator, transducer, and effector proteins. Viruses modulate downstream signaling by activating or inhibiting mediators, transducers, and effectors in the DDR. (3) Cellular consequences occur in response to recognition and signaling in the form of DNA repair, cell cycle checkpoints, chromatin remodeling, and transcriptional changes. Viruses can modify repair pathways, elicit specific cell cycle arrest, alter chromatin organization, and induce transcriptional changes (4) Innate immunity is directly tied to these processes as DDR factors can elicit antiviral activity. Together, these four pillars represent the interplay between viruses and the DDR and exemplify how the DDR and innate immunity are directly interconnected and central to viral replication.
Overview of the DNA damage response (DDR)
The first step in activation of the DDR is induction of DNA damage. DNA damage occurs through endogenous pathways, such as DNA replication errors and reactive oxygen species generated during cellular metabolism, as well as exogenous factors, such as ultraviolet and ionizing irradiation, chemical mutagens, and viral replication. These genotoxic stresses can result in double-strand DNA breaks (DSBs), single-strand DNA breaks (SSBs), and single-base modifications such as mismatched bases, DNA adducts, or intra-strand crosslinks. Depending upon the type of damage, specific protein sensors are responsible for recognizing damaged DNA and initiating the DDR signaling. For the purpose of this review, we will focus on sensing and signaling associated with DSBs and SSBs (Figure 2). DSBs are primarily recognized by the MRE11, Rad50, NBS1 (MRN) complex, leading to ATM activation and recruitment to sites of genotoxic stress. Active ATM phosphorylates various downstream effector proteins including histone variant γH2AX, CHK2, and 53PB1.4–6 Alternatively, DSBs can be recognized by the DNA-PK holoenzyme (Ku70, Ku80, and DNA-PKcs) to be repaired by non-homologous end joining (NHEJ), a more error-prone process than homologous recombination (HR) repair and primarily occurs in the G1 phase of the cell cycle.4–6 SSBs are sensed by RPA; when bound to ssDNA, RPA activates ATR, which stimulates downstream signaling proteins such as CHK1.4–6 Depending on the type and severity of the lesion, activation of the DDR results in various cellular outcomes such as DNA repair, cell cycle arrest, chromatin dynamics, and transcriptional changes.
Figure 2. The DDR senses, signals, and responds to aberrant DNA through three primary pathways, ATM, ATR, and DNA-PK, that are modulated by viruses.

(1) The DDR is a protein signaling cascade that maintains genome integrity. The DDR consists of sensors, which recognize specific DNA lesions, mediators and transducers that transmit this signal of damaged DNA, and effectors, which directly execute a cellular response. While many of these pathways are interconnected, in general, the ATR pathway is activated in response to SSBs, and ATM and DNA-PK pathways are activated in response to DSBs. DDR signaling induces cellular consequences, including DNA repair, cell cycle arrest, chromatin dynamics, and transcriptional changes. Shown are representative DDR proteins, pathways, and cellular consequences that are highlighted in this review with the exception of apoptosis and senescence. (2) Viruses have developed several mechanisms to activate, inhibit, or degrade various parts of these core signaling pathways. Modulation of these pathways ultimately leads to several cellular consequences which are beneficial to the viral lifecycle.
I. Viral manipulation of the DDR
In the following section, we will discuss examples of how diverse viruses modulate all steps of the DDR, including induction of DNA damage, recognition by damage sensors, signaling via mediator, transducer, and effector proteins, and cellular consequences of the DDR (Figure 2 and Table 1). We have focused on specific examples which we hope will convey three main points: (1) modulation of the DDR is conserved by diverse viruses, regardless of viral genome type or location of replication; (2) the DDR both enhances and inhibits viral replication; (3) specific proteins as well as DDR signaling pathways play important roles in viral replication. In addition, while not explicit to this section, we will begin to highlight how the DDR and innate immunity are directly linked, which is further explored in the second part of this review.
Table 1.
A summary of various viruses and the cellular consequences caused by viral modulation of the DDR.
| Genome Type | Virus | Viral Proteins | Damage | Signaling | Repair | Cell Cycle | References |
|---|---|---|---|---|---|---|---|
|
| |||||||
| (+) RNA | CoV | CoV-N, nsp13, p125 | Stalled Forks | ATR | N/S | G0/G1, G2/M | 69–72,183 |
| (−) RNA | IAV | NS1 | DSB | ATM, DNA-PK | Modulate MMR | G0/G1 | 56,57 |
| dsRNA | Rotavirus | NSP2, NSP5 | N/S | ATM | N/S | G2 | 36,55 |
| Retro | HIV | Tat, Vif, Vpr | SSB, DSB | ATR ATM | Repress HR | G1, G2/M | 13–15,61,76,77,79 |
| Retro | HTLV | Tax, p30 | DSB | ATM | Repress HR, Promote NHEJ | G1, G2 | 10,73–75 |
| DNA | HPV | E1, E2, E6, E7 | DSB, Stalled forks | ATM, ATR | Repress HR | G2 | 50–52,58 |
| DNA | EBV | LMP-1, EBNA3C, ZEBRA | DSB | ATM | N/S | G2/M | 85,95,173,184–189 |
| DNA | Adenovirus | E4, E1a, E1b, EIB55K, E4orf6 | N/S | ATM, ATR, DNA-PK | Repress NHEJ | Dysregulation | 44,53,54,65,94 |
Damage, signaling, cell cycle, and DNA repair are four major phenotypes central to characterizing the ability of viruses to modulate the DDR that we recommend the field focus on moving forward. N/S indicates not shown.
Induction and recognition of host DNA damage
Viruses induce host DNA damage
Many examples exist that demonstrate induction of DNA damage during viral replication. Simian Virus 40 (SV40), a dsDNA virus, induces DNA damage via the large T antigen,7 and Human adenovirus type 12 (Ad12) induces chromosomal aberrations in human embryonic kidney cells (HEK).8 Influenza A (IAV) subtype H3N2, a segmented (−) RNA virus that replicates in the cytoplasm and the nucleus, causes DNA damage in leukocytes early during infection.9 Human T-lymphotropic virus type 1 (HTLV-1), a retrovirus, induces DSBs during DNA replication through Tax.10 While retroviruses may induce DNA damage through the process of integration,11,12 it is becoming more apparent that retroviruses also induce DNA damage independently of integration, which we will describe throughout this review. However, what remains unclear for many of these viruses is how DNA damage occurs, whether viral-induced DNA damage is sensed and signaled by canonical cellular DDR pathways, and what role induction of DNA damage plays in viral replication and disease pathogenesis.
One example of a viral protein that induces DNA damage is the lentiviral protein Vpr. Human Immunodeficiency Virus 1 (HIV-1) Vpr induces both SSBs and DSBs, independent of other lentiviral proteins.13–15 While Vpr localizes to chromatin and is reported to bind DNA,16–18 it does not display any nuclease activity, suggesting Vpr may induce DNA damage through an indirect mechanism.14 One possibility is that Vpr induces DNA damage indirectly by binding to chromatin and inhibiting DNA replication,19 leading to DNA damage following replication fork collapse. However, Li et al. showed that Vpr causes DNA damage independently of DNA replication stalling.15 Another leading hypothesis is that Vpr induces DNA damage as a consequence of degradation of a DNA repair protein. Vpr recruits the host Cul4ADCAF1 ubiquitin ligase complex and interacts with many host DDR proteins – including UNG2,20,21 HLTF,22,23 SLX4 complex proteins MUS81 and EME1,24,25 EXO1,26 TET2,27 MCM10,28 hHR23A,29 and SAMHD130,31 – yet degradation of most of these proteins has not been shown to be required for induction of DNA damage likely because the function of Vpr is complex and induces proteomic changes across the entire cellular landscape.15,32,33 Thus, like many other viral proteins, it remains unclear exactly how DNA damage is induced by Vpr, and further exploration will better inform us why viruses induce DNA damage.
One of the problems we face in the viral DDR field is that many of the central phenotypes of viral DDR modulation have not been tested directly or with methods that are easily reproducible. For example, induction of DNA damage has often been identified through detection of γH2AX activation rather than probing for DNA damage directly. Utilizing γH2AX in lieu of detecting DSBs or SSBs is problematic because activation of γH2AX is not necessarily a direct indicator of DSBs or SSBs, and γH2AX could potentially be activated by viruses in the absence of DNA damage. To ameliorate this, we recommend that the virologists move toward directly testing for DNA damage through more specific DNA damage assays (Box 1).
Box 1. Methods.
Using assays that directly test for DNA damage or repair and are easily reproduced is a crucial step towards synthesizing the mechanisms by which viruses modulate the DDR. For example, γH2AX is often used as a proxy of DNA damage. This is problematic, since γH2AX activation can occur in the absence of DSBs or SSBs. Furthermore, γH2AX activation fails to distinguish between induction of DSBs or SSBs, leaving the type of DNA damage induced by the virus unknown. We recommend using assays that directly test for DNA damage, for example, the comet assay and pulsed-field gel electrophoresis (PFGE) that directly detect DSBs and SSBs. The comet assay quantifies DNA damage at single-cell resolution. Cells are embedded in an agarose-coated glass slide, permeabilized, and gel electrophoresed. Fragments of DNA that contain SSBs or DSBs are separated from the core of the cell and create a “comet tail.” The amount of DNA damage per cell is quantified by measuring the percent of DNA in the comet tail.162 Similarly, the PFGE assay is an electrophoresis-based DNA fractionation method to detect DSBs. Cells are embedded in an agarose gel, lysed, and undergo PFGE, leading small DNA fragments to migrate in the agarose gel. The total quantity of damaged DNA is determined along with the molecular sizes of the fragmented DNA.163 To directly test for DNA repair, we recommend using I-Sce1-based reporter assays, such as the DR-GFP assay for HR that are amendable to various cell types. Two incomplete GFP cassettes are stably integrated into the genome of cell lines such as U2OS. The first GFP gene contains an I-SceI endonuclease cut site, and the second contains a truncated GFP sequence. Transient expression of I-SceI in HR- or NEHJ-proficient cells induces a DSB in the first GFP gene that once repaired results in GFP+ cells, which are then detected and quantified using flow cytometry. Additional GFP reporter systems have been developed to assay single strand annealing and alternative end joining DNA repair.164–167 While CRISPR-based assays are starting to make headway due to their flexibility in different cellular systems,168–170 assays such as the DR-GFP are conventional assays in the field.
Viruses modulate DDR sensors
Subsequent to induction of DNA damage, viruses also modulate the primary sensors of this damage, including the MRN complex, RPA, and Ku70/80 (Figure 2). The MRN complex, which is a major sensor of DSBs, has been shown to inhibit replication of many diverse viruses.34–36 Thus, many viruses inhibit MRN. For DNA viruses such as adenoviruses, the MRN complex inhibits viral replication primarily by impairing viral DNA replication. To overcome this inhibition, Ad5 employs multiple E proteins to both relocalize and degrade components of the MRN complex.35,37,38 Interestingly, not all Ad serotypes can overcome the MRN complex,39–41 indicating differences in the evolution of MRN antagonism. Another dsDNA virus, the herpesvirus Kaposi’s Sarcoma-associated Herpesvirus (KSHV), antagonizes MRN through the viral LANA protein to block innate immune inhibition of viral replication and to support lytic reactivation.42 Similar to adenoviral E proteins, LANA facilitates the relocation of the MRN complex to the cytoplasm. Additionally, RNA viruses such as rotavirus antagonize MRN by relocalizing the complex to the cytoplasm via viral proteins NSP2 and NSP5,36 and the retrovirus HTLV-1 p30 directly binds to MRN components Rad50 and NBS1 to sequester and inhibit MRN complex formation.43 MRN antagonism through sequestration and/or relocalization is conserved among diverse viruses, suggesting that evading damage detection by MRN is a strategy beneficial for productive infection.
Viruses also modulate the heterodimeric SSB sensor RPA (composed of RPA70, RPA32, and RPA14). However, unlike MRN antagonism, viruses primarily activate RPA – indicating that RPA enhances viral replication. For example, the Ad5 and Ad12 E1B-55K protein directly interacts with the host E1B-AP5 protein, which binds to the RPA component RPA32 in adenovirus replication centers. This is essential for ATR-dependent phosphorylation of RPA32, suggesting Ad5 and 12 regulate the ATR pathway through direct modulation of RPA phosphorylation.44 For HIV-1, Zimmerman et al. showed that Vpr is responsible for inducing activated RPA foci in primary human CD4+ lymphocytes.19 However, Vpr does not colocalize with RPA32 foci, suggesting Vpr may indirectly modulate RPA activity.15 Thus, while less well defined than MRN antagonism, some viruses have evolved to activate RPA through direct and indirect mechanisms.
Finally, viruses antagonize the DSB sensor Ku70/80 complex, which is required for NHEJ-mediated DNA repair. For example, HTLV-1 transcriptionally silences Ku80 expression, inhibiting DNA-PK and innate immune activation.45 Antagonism of Ku70/80 is important to the viral lifecycle as Ku70 directly recognizes a HTLV-1 reverse transcription intermediate (ssDNA90) and stimulates type 1 IFN and cytokine production, which together limit HTLV-1 infection before retroviral integration.46 Therefore, Ku70/80 complex antagonism may be necessary to overcome innate immune sensing and to promote viral replication.
DDR signaling
Downstream of sensing DNA damage, there is a vast signaling cascade consisting of mediator, transducer, and effector proteins (Figure 2). Despite differences in genome type and where they replicate in the cell, DNA, RNA, and retroviruses have evolved several mechanisms to modulate the DDR effectively. Although virus-induced DDR signaling is broadly conserved across viruses, the viral classification does not necessarily correlate with the proteins and pathways modulated. Here we will discuss and exemplify the three primary mechanisms that viruses use to engage DDR signaling: activation, inhibition, and degradation (Figure 2).
Human Papilloma Virus (HPV) strains modulate many aspects of the DDR associated with ATR and ATM, predominantly through the early viral proteins E1, E2, E6, and E7. Most notable is the capacity of E6 and E7 to directly interact with p53 and Rb, two important regulator proteins in DDR signalling,47–49 respectively.50,51 By inhibiting p53, E6 directly affects the ATR and ATM pathways, which in turn alters cellular processes such as cell cycle, DNA repair, and transcription. In addition to Rb binding, Moody and Laimins showed that high-risk HPV-37 E7 directly interacts with ATM leading to phosphorylation of Ser1981, causing activation and further downstream phosphorylation of CHK2. Moreover, they observed phosphorylation of CHK1 by E7, which is typically associated with ATR signaling, further suggesting a potential role for ATR in HPV replication.52 Many of these signaling pathways are activated directly by viruses through alternative mechanisms and are important for viral replication. However, whether ATM and ATR are both activated by diverse HPV subtypes, whether this activation occurs in conjunction with inhibition of either p53 or Rb, and whether this is dependent on antagonism of MRN remains to be studied.
As previously discussed, adenoviruses impair ATM signaling through sequestration of the MRN complex. However, adenoviruses also directly impair DNA-PK signaling, highlighting the necessity to target multiple arms of the DDR. Because all adenoviruses encode a linear double-stranded genome, they are particularly vulnerable to DNA-PK, which functions by re-ligating DSBs with exposed ends. As a response, adenoviruses disable the DNA-PK pathway by proteasome mediated degradation of DNA ligase IV via the interaction of E4 and E1b proteins with the host Cul5 ubiquitin ligase complex.53,54 Consequently, this allows the virus to replicate efficiently without being antagonized by host repair machinery, which acts as an antiviral defense mechanism and highlights the capacity of canonical DDR associated proteins to exhibit antiviral innate immune functions (Figure 3).
Figure 3. Innate Immune sensors and DDR signaling factors are two sides of the same coin.

Innate immune sensors and DDR proteins and pathways have been canonically attributed to either innate immunity or DDR signaling, but there are multiple examples of how proteins being multifunctional and involved in both processes. (1) cGAS can function in the nucleus and interacts directly with chromatin, and (2) ATM is not limited to the nucleus and can relocalize to the cytosol. (3) Both cGAS and ATM can activate NF-κB signaling. (4) DDR factors involved in NHEJ repair, such as DNA-PK, can have direct antiviral activity and inhibit adenovirus, (5) and innate immune factors such as SAMHD1, which inhibit HIV, are also involved in nuclear DDR signaling.
The viral lifecycle of many RNA viruses is primarily cytosolic. Despite this, RNA viruses still take advantage of the DDR machinery that largely reside and function in the nucleus. Rotavirus, a double-stranded RNA virus, is an interesting example of an RNA virus that modulates the DDR. The viral proteins NSP2 and NSP5 noncanonically activate ATM signaling independent of DNA damage and γH2AX activation and further relocalize ATM, CHK2, and the MRN complex from the nucleus to the cytoplasm.36 Strikingly, ATM and CHK2 only interact with NSP5 in the presence of replicating viral genomes and inhibition of the ATM pathway reduces viral replication, suggesting that activation of these signaling pathways is important for viral genome replication.55 Orthomyxoviruses are also of particular interest as they consist of a segmented (−) RNA genome that, unlike many RNA viruses, is shuttled to the nucleus for genome replication and transcription. As such, these viruses also encode nuclear viral proteins to modulate the host DDR effectively. Specifically, the IAV viral protein NS1 suppresses RhoA and pRb signaling, directly activating the ATM signaling cascade.56 In addition to these pathways, IAV infection modulates the protein abundance of various fundamental DDR proteins, such as Ku70, Ku80, Rad51, γH2AX, and PCNA, all of which are critical for ATM, ATR, and DNA-PK signaling.57 Altogether, this exemplifies the evolutionary importance of modulating nuclear DDR factors for all viruses and could suggest that engagement of the DDR drove nuclear replication of some RNA viruses such as orthomyxoviruses and retroviruses. Despite cellular localization of viral replication, viruses require host DDR factors that they do not encode to replicate and thus evolving to activate the DDR through both canonical and noncanonical mechanisms is crucial for viral replication.
Cellular consequences of DDR
Depending on the type and severity of the genomic lesion, activation of the DDR results in a myriad of cellular consequences, including but not limited to DNA repair, cell cycle arrest, chromatin dynamics, and transcriptional changes (Figure 2). In this section, we will highlight how diverse viruses utilize common mechanisms to alter the cellular consequences of the DDR. We will specifically focus on how DNA, RNA, and retroviruses dysregulate DNA repair, promote cell cycle arrest, confer changes in chromatin organization, and induce transcriptional changes.
Repair
One of the major consequences of modulating the DDR is the disruption of the five primary repair pathways: BER, NER, and MMR, which repair single-strand lesions, and HR and NHEJ, which repair DSBs. Disruption of DNA repair has been observed for many of the viruses that we have discussed thus far. Despite how much is known about viral modulation of DDR signaling, repair as a cellular consequence remains poorly understood. HR is one of the major repair pathways a cell utilizes extensively during late S and G2 phases of the cell cycle and can be activated in response to DSBs, primarily via ATM signaling. Fittingly, many viruses that regulate ATM signaling also regulate HR. For example, HPV represses HR efficiency by 50–60% through the recruitment of a variety of DNA repair host factors such as Rad51, RPA70, BRCA1, and BRCA258 away from chromatin and relocalization to the viral genome.59 Consequently, these cells are more sensitive to exogenous genotoxic stress.60 The role of DNA repair pathways in HIV infection is not well understood. In one system, HT1080 cells were transfected with pBHRF, a plasmid vector used to measure HR, in the presence or absence of transfected Vpr to assess HR repair of truncated GFP. In the presence of Vpr, GFP expression increased, suggesting Vpr enhances HR.61 Using the DR-GFP assay to assess repair efficiency (described in Box 1), HR and NHEJ have been shown to be repressed in U2OS cells expressing HIV-1 or HIV-2 Vpr.15 Since discrepancies remain for the effects of Vpr on HR, it is crucial that the field uses similar systems and assays, such as the DR-GFP assay, to create reproducible and comparable data as discussed further in Box 1. This will also help to determine whether functions such as repression or activation of HR are directly beneficial to viral replication or a consequence of redistributing host DDR factors.
In addition to HR, other repair mechanisms can also play an important role in viral replication. As previously discussed, adenoviruses broadly inhibit the DNA-PK pathway by disrupting DNA ligase IV activity via proteasomal degradation, leading to the downregulation of NHEJ, which affects processes such as V(D)J recombination.54 Though the role of DNA repair in RNA viruses remains understudied, it has been proposed that DNA repair is exploited during RNA virus infections. For example, IAV modulates and exploits MMR to promote cell survival during infection. An MMR activity assay, which utilizes a mismatch start codon on a luciferase expression plasmid, revealed that maintaining MMR activity is important for the IAV viral life cycle.62 Strikingly, unlike HR or NHEJ, MMR activity leads to decreased transcription of antiviral innate immune factors, suggesting that affecting this particular DNA repair pathway has the additional cellular effect of dampening the innate immune response that would otherwise inhibit viral replication.
Cell cycle
Many viruses utilize the DDR to inhibit or activate cell cycle progression to facilitate an environment conducive to viral replication. Our current understanding is that certain cell cycle phases, such as S-phase, can promote viral replication, whereas the roles of others, such as G1 or G2/M, are still less clear. Here we will highlight different strategies viruses have evolved to both inhibit and promote cell cycle progression to benefit viral replication.
dsDNA viruses are the textbook example of cell cycle control by a virus, as they require a cell to be in S-phase in order to replicate their genomes.63 This is because dsDNA viruses utilize much of the same machinery as the host to replicate DNA, including cellular DNA replication proteins and dNTPs. To achieve this, almost all dsDNA viruses encode early proteins that directly inhibit the master cell cycle regulators Rb and p5347–49 through degradation, relocalization, and/or sequestration.64,65 Some examples include HPV E6 and E7 proteins, adenovirus E1A and E1B proteins, and SV40 large T antigen, and has been extensively reviewed elsewhere.3,66–68 By studying how dsDNA viruses regulate S-phase, we have not only learned about mechanisms of viral replication but have also uncovered many molecular mechanisms underlying cell cycle regulation and the cellular consequences of dysregulation, such as cancer. Thus, viruses have been an instrumental tool in understanding viral and host biology.
Unlike the aforementioned DNA viruses that primarily push cells into a single cell cycle stage, coronaviruses represent a single family of RNA viruses that have all evolved to differentially regulate the cell cycle and display a range of cell cycle phenotypes. This is accomplished through an assortment of viral proteins, such as CoV-N, nsp13, p28, and ORF3/M, which converge on inhibiting cyclin-CDK complexes or upstream signaling cascades, such as p53, to induce cell cycle arrest. Consistent with the different viral proteins, the type of arrest induced varies between G0/G1, S, and G2/M.69–72 Despite these differences, induction of cell cycle arrest is conserved among coronaviruses and allows viruses to exploit host resources, such as translation and replication factors, that are essential for viral replication but are not encoded by the viral genome.
Many retroviruses also alter cell cycle; though the effects on cell cycle progression and viral replication seem to be distinct. For example, HTLV-1 infection allows cells to bypass the G1/S checkpoint despite DNA damage.73 This is regulated by the interaction of HTLV-1 Tax with the cellular phosphatase Wip1, which dephosphorylates γH2AX and RPA to bypass the DDR-initiated G1/S checkpoint.73 Interestingly, Tax has more than one function and also mediates G2 accumulation through the direct binding and activation of CHK2 independent of ATM.74,75 Primate lentiviruses induce arrest, with at least three HIV-1 proteins implicated in a G1 (Tat) or G2/M (Vif and Vpr) arrest.76–79 The primary role of cell cycle arrest in HIV-1 replication is unclear, but G2/M arrest has been proposed to promote viral expression80 and/or prevent nuclear breakdown to exploit nuclear factors in cycling T cells. Lentiviruses also have the ability to infect non-dividing cells, such as macrophages and dendritic cells. While at least one study has shown that prevention of cell cycle progression into mitosis in monocyte-derived dendritic cells is important for LTR-mediated viral transcription,81 it will be important for the field to directly address the role of activating cell cycle-associated pathways in noncycling cells.
Chromatin dynamics
Chromatin bound to damaged DNA must reorganize to allow DDR proteins to access damaged DNA and facilitate repair. Histone proteins bound to damaged DNA undergo post-translational modifications (PTMs), such as methylation, acetylation, phosphorylation, and ubiquitylation. This alters chromatin structure, DNA repair, and the local transcriptional environment. Thus, many viruses directly target histone modifying proteins to influence the availability and abundance of nuclear factors and ultimately enhance viral replication. Some of the more widely conserved viral targets, which we will specifically discuss here, are the ubiquitin ligase proteins RNF8 and RNF168 and the acetyltransferase Tip60.
RNF8 and RNF168 are ubiquitin-protein ligases that play key roles in DNA damage signaling by catalyzing and amplifying ubiquitylation of histones H2A and H2AX to promote the recruitment of DNA repair proteins at DSBs.82 Viruses inhibit RNF8 and RNF168 through several diverse mechanisms, including degradation or relocation of RNF8 and limiting the recruitment of DDR proteins to sites of damage. For example, HSV-1 ICP0 degrades RNF8 and RNF168, causing the loss of H2A ubiquitylation and DNA repair factor recruitment to DNA damage sites; thus, inhibiting DDR signaling.83 Similarly, HPV E7 directly binds to and inhibits RNF8, which again limits the recruitment of 53PB1 to radiation-induced damage sites and increases repair by HR.84 The EBV immediate-early protein BZLF1/ZEBRA similarly antagonizes RNF8 by relocating RNF8 and 53BP1 away from sites of DNA damage and consequently inhibiting DNA damage repair.85 HTLV-1 Tax relocalizing RNF8 from the nucleus to the cytoplasm stimulates the DDR and induces assembly of K63-pUb chains that also activate NF-κB.86 Together, this exemplifies the central role ubiquitylation plays in viral modulation of the host DDR to alter the availability of DDR factors.
Viruses also alter the chromatin environment by modulating the host acetyltransferase Tip60, which is a component of the NuA4 complex that acetylates histones to regulate gene expression and DNA repair.87 Tip60 also directly regulates DNA repair by acetylation and activation of ATM, independent of NuA4.88 Tip60 was first identified through its interaction with the HIV-1 transcriptional activator Tat.89 While the precise role of the Tat-Tip60 interaction in HIV-1 replication remains unclear,90–92 binding of HTLV-1 p30II to Tip60 promotes acetylation, chromatin remodeling, and transcription of c-Myc target genes,93 suggesting this could be a conserved and important retroviral-host interaction.
In addition to altering the chromatin environment, Tip60 inhibits gene expression of several dsDNA viruses, including adenoviruses,94 herpesviruses,95–97 papillomaviruses,98–101 and the hepadnavirus HBV,102 and thus, DNA viruses have evolved diverse mechanisms to antagonize Tip60. For example, adenovirus antagonizes Tip60 through the viral EIB55K and E4orf6. Both EIB55K and E4orf6 bind to Tip60 during infection and target it for proteasome-mediated degradation, causing cellular chromatin inaccessibility and promoting viral early gene transcription.94 Recently, Tip60 was indicated to be upregulated in response to IAV infection and to activate type I IFN.103 It remains to be seen whether Tip60 and other histone modifying proteins may have additional roles in response to viral infection.
Transcriptional changes
Another major consequence of DNA damage is modulation of the cellular transcriptome. Specifically, DNA damage limits global transcription by inhibiting RNA polymerase II104 and promoting activation of specific transcriptional pathways, such as NF-κB. Activation of NF-κB by DNA damage is dependent on the ATM-NEMO pathway and upregulates NF-κB-regulated genes important for facilitating cell survival by inhibiting apoptosis and mediating DNA repair105–107 (reviewed in 108).
Many viruses induce DNA damage causing transcriptional changes that benefit viral replication and are often linked to NF-κB.42,109–112 For example, HPV regulates transcription initiation by recruiting NF-κB through the viral helicase E1. Activation of NF-κB leads to the destabilization of E1, establishing a negative feedback loop to regulate E1-dependent genome amplification and NF-κB transcriptional changes.113 Similarly, during HTLV-1 and HIV-1 co-infection, HTLV-1 Tax can regulate transcription initiation by facilitating the recruitment of NF-κB to the unintegrated HIV-1 LTR. Mechanistically, HTLV-1 Tax promotes the recruitment of the NF-κB subunits RelA and RelB to the HIV-1 LTR, which induces viral gene expression and facilitates viral replication.114 HTLV-1 can also activate NF-κB through Tax-independent mechanisms, which promote survival and proliferation of HTLV-1 infected cells by upregulating genes responsible for proliferation and clonal expansion.115 Interestingly, transcriptional changes induced by DNA damage enhance viral gene expression and promote viral replication. For example, DNA damage via ultraviolet light or mitomycin C enhances transcription of the HIV-1 LTR,116 further suggesting that DNA damage can also alter viral transcription (see Box 2 for more information on the DDR and reactivation of latent viral gene expression).
Box 2. Latency and the DDR.
Viral latency is a reversible state characterized by a long-term infection where no new viral particle is formed. As latent viruses are often dormant and sometimes silent, these reservoirs make it difficult to clear an infection, allowing for latency reversal and viral persistence. The two viral families most often associated with latency are herpesviruses and retroviruses. As with active infections, latent viral infections can also cause DNA damage. One well studied example is EBV, which still expresses low levels of some viral proteins during latency, including nuclear antigen (EBNA) and membrane protein (LMP).171 Combined expression of these proteins can lead to an increase of reactive oxygen species that can damage DNA.172,173 Latently expressed EBNA and LMP also impair the DNA damage response through modulating the autophagic response, leading to accumulation of DNA damage without cell death173 and contributing to the oncogenic potential of EBV. Cells infected with latent HIV are more sensitive to DNA damaging agents,174,175 which may provide a novel approach to selectively purge latently infected cells. Another key mechanism linking latency and DNA damage is the potential for latency reversal through DNA damage. For example, ultraviolet irradiation enhances reactivation of many viruses, including HIV-1 in cell culture and animal models,116,176 most probably by exposing transcriptionally repressed regions to host transcription machinery through changes in epigenetic status of the latent viral genome.177,178 This suggests that modulating DNA damage can regulate viral replication in latent cells. By concentrating more on the ways DNA damage and latency are related, we could further understand both the effects of a latent infection and how to clear a latent reservoir.
II. Crosstalk between the DDR and innate immunity
Both the DDR and the innate immune system are signaling cascades that function to sense, signal, and respond to aberrant nucleic acids. Once considered to be independent cellular functions, evidence now suggests that they may not be mutually exclusive processes, and there is more crosstalk than previously considered (Figure 3). For example, recent research indicates that the major innate immune DNA sensor cGAS can also sense perturbations and disruptions of genome maintenance and DNA repair and potentially even inhibit HR (Figure 3).117–121 In this section, we examine the interplay between innate immunity and the DDR and the “role reversal” of a few proteins canonically believed to either play roles in innate immunity or the DDR. We will highlight recent data supporting the crosstalk between the DDR and innate immunity to make explicit the connections between the two systems. Since we will not be discussing the innate immune response to viral infection, please see the suggested reviews for more information.122,123
SAMHD1
SAMHD1 sits at the intersection of innate immunity and the DDR. SAMHD1 is a dNTP hydrolase that depletes intracellular dNTP pools and is an important antiviral restriction factor against primate lentiviruses such as HIV-1 (Figure 3).124,125 SAMHD1 also shows evolutionary signatures of long-standing genetic conflict, called positive selection, consistent with an important conserved role in antagonizing viral replication (see Box 3 for more information on the rapid evolution of DDR genes).30,31,126
Box 3. DDR genes under positive selection.
DNA damage signaling and repair pathways critically maintain genome integrity and exhibit high evolutionary conservation across eukaryotes. Despite this, the sequences of many genes that comprise these pathways are marked by signatures of rapid evolution, called positive selection (PS). PS is often the consequence of long-standing evolutionary conflict and can be indicative of an important role in viral replication.179 These signatures can be found in genes encoding proteins involved in specific DDR pathways, including HR and NHEJ, which have been shaped by recurrent PS.180 Crystal structures of primate NHEJ factors XRCC4, Nbs1 and Polk reveal PS sites located exclusively on exposed protein surfaces, supporting the idea that binding of viral proteins is driving the rapid evolution of residues at virus-host interfaces. Yet, due to the critical nature of NHEJ factors in cell survival, the functions of these genes are evolutionarily constrained, and sites of PS don’t appear to alter their primary roles in DNA repair. In some cases, however, recurrent PS may lead to altered functions of host genes, even in genes in which mutations often lead to cancer. For instance, BRCA1 and BRCA2 show signatures of PS,181 though whether they have been driven by viruses has yet to be determined. Because BRCA1/2 are responsible for most hereditary forms of breast and ovarian cancer,182 one would expect high conservation in the sequences of these essential genes. Thus far, these signatures have resulted in nonsynonymous point mutations in BRCA1/2 rather than the premature stop codons and frameshifts immediately associated with cancers. Yet, it is possible that the subtle effects of these mutations on BRCA1/2 function and cancer-risk may take years to be realized. Eventually, this recurrent PS may result in antagonistic pleiotropy, where positively-selected-for BRCA1/2 residues would favor viral resistance at the expense of an elevated risk of cancer.181
SAMHD1 plays an important role in the DDR to maintain genome integrity. Patients with either Aicardi-Goutières syndrome, a severe autoimmune disorder, or chronic lymphoid leukemia that also have SAMHD1 loss-of-function mutations have increased levels of dNTPs and DNA damage, suggesting that SAMHD1 engages the DDR. This is consistent with previous research demonstrating dysregulated dNTP levels can activate the DDR through replication stress.127–129 Overexpressed HA-tagged SAMHD1 has been shown to colocalize with 53BP1 even at steady state; though the number and size of the foci increase after treatment with the DNA damaging agent camptothecin.128 SAMHD1 can mediate HR repair through recruiting and interacting with CtIP at sites of DNA damage and promoting DNA end resection.130 Coquel et al. showed SAMHD1 physically interacting with MRE11 at stalled replication forks and stimulating the exonuclease activity of MRE11 to degrade nascent DNA. ATR–CHK1 is activated, restarting the replication forks. Without SAMHD1 acting at stalled replication forks, ssDNA fragments are released into the cytosol, where they activate the cGAS-STING pathway.131 Taken together, SAMHD1 is recruited to sites of DNA damage, and loss of SAMHD1 leads to replication fork stalling, demonstrating SAMHD1 has a direct role in genome maintenance.
SAMHD1 is activated in response to specific DNA damage and inhibits HIV-1 infectivity. Induction of DSBs through the use of neocarzinostatin, etoposide, or camptothecin activates SAMHD1 in monocyte-derived macrophages and blocks HIV-1 infection.132,133 However, stimulating the DDR with ultraviolet light did not activate SAMHD1, suggesting it is DSBs that are important for SAMHD1 activation.133 Interestingly, treatment with camptothecin greatly reduced infectivity, and cells had better survival in comparison to etoposide-treated cells. This suggests DNA damaging agents block HIV-1 infectivity with different efficiencies, perhaps due to the level of DNA damage induced. The authors proposed that DNA damage activates SAMHD1 causing a post-RT block and inhibition of HIV-1 infectivity.132 Neocarzinostatin, etoposide, camptothecin and ultraviolet light all stimulate γH2AX, suggesting activation of γH2AX is not sufficient to activate SAMHD1.133
ATM
Ataxia–Telangiectasia (AT) syndrome patients display worsening movement coordination, a weakened immune system, and an increased risk of cancer. ATM is also a key protein in the canonical NF-κB response to genotoxic stress (Figure 3),134–136 and recent work has expanded upon the role ATM plays in innate immunity. In irradiated mouse macrophages, ATM stimulates an IFN response in a STING-independent, IRF1-dependent manner, in addition to activating NF-κB.137 ATM has also been shown to play a role in an IFI16-STING-IRF3 signaling cascade in response to etoposide treatment. In that model, ATM phosphorylates p53 which then associates with IFI16 and STING in the cytoplasm.138
The expanding role of ATM in innate immunity during microbial infections is under active investigation. Currently, the evidence is murky, with most of the work focusing on antibacterial responses. Though this does not specifically inform the role of ATM during antiviral immune responses, by understanding the role of ATM during the antibacterial immune response, we can learn how ATM generally interacts with and within innate immunity. Conflicting publications have shown that the loss of ATM can either hinder or enhance antimicrobial responses. Härtlova et al. saw enhanced antiviral and antibacterial responses in cells from patients with AT and ATM knockout mice. Unrepaired DNA lesions in ATM deficient cells activates STING and the production of type I IFNs.139 However, in their follow-up paper, ATM knockout mice were more susceptible to pulmonary bacterial infections. They concluded the loss of ATM leads to ROS production, impairing some innate immune sensors.140 In another publication, murine macrophages deficient in both ATM and DNA-PK had reduced cytokine production when infected with Listeria monocytogenes. However, they only included data for macrophages deficient in both ATM and DNA-PK or only DNA-PK, so the individual role of ATM in this system is unclear.141 Conversely, Härtlova et al. infected ATM knockout mice with L. monocytogenes and still saw an increase in type I IFNs.139 The differences between all of these publications could be due to using assays specific for certain innate immune pathways versus others, or the experimental system such as in vitro macrophages vs. in vivo mouse models. Not only does more work need to be done to clarify what role ATM might play in an innate immune response, but also specifically the importance of ATM in the response to viral pathogens.
MRE11, MUS81, RAD51, and RPA
Within the DDR there are sensors, mediators, transducers, and effectors that play a role in activating or repressing innate immunity. MRE11 has been shown to localize to the cytoplasm to detect dsDNA and activate STING dependent type 1 IFN expression. HEK293 cells treated with IFN stimulatory DNA or ultraviolet light accumulate activated ATM, suggesting the DDR is activated in response to extracellular DNA and DNA damage. Activation of type I IFN genes were detected in response to treatment with the extracellular DNA and decreased in a MRE11 knock-down. The MRE11 binding partners RAD50 and NSB1 are required to sense DSBs, yet neither were required to respond to HSV-1 or the bacterial pathogen L. monocytogenes. Thus, the working model suggests that MRE11 alone may initiate a type I IFN response downstream of cell-intrinsic damage.142
MUS81 is a DNA repair endonuclease that forms a complex with EME1, SLX4 and SLX1 and is important for DNA replication through its regulation of replication fork progression.143,144 HIV-1 downregulates Mus81 through recruitment of the Cul4ADCAF1 ubiquitin ligase complex,24,25 suggesting a potential role for this host DDR protein in innate immunity. Additionally, it has been shown that MUS81 activity leads to the accumulation of cytosolic dsDNA fragments in prostate cancer cells. These fragments are recognized by the STING pathway leading to type I IFN production, but they did not investigate which specific dsDNA sensor is required. Some data indicate that the immune response stimulated by MUS81-STING is required for rejection of the tumor cells when injected into mice. This is very preliminary and needs to be followed-up on more thoroughly.145 However, the role STING plays in both the development of but also the potential treatment of cancer is a growing area of research.146
While some DDR factors sense and activate an antiviral response, other DDR factors can inhibit aberrant activation independent of viral infection. RAD51 and RPA are involved in DNA repair following damage. Two publications have shown them to be important for protecting against leakage of ssDNA into the cytosol, which could stimulate an innate immune response in the absence of viral infection. Depleting just one is sufficient to cause increased levels of ssDNA fragments in the cytosol and IFNb production via the cGAS-STING pathway. Thus, implying that RAD51 and RPA protect a cell from initiating an inflammatory response to self-DNA.147,148
DNA-PK
As previously discussed, DNA-PK is a heterotrimeric protein complex that is an essential mediator in the cellular response to DNA damage and is targeted by viruses. Using pulldowns in HEK293 and HEK293T cells, two groups identified that DNA-PK can bind DNA in the cytoplasm, and depending upon the cell type used, initiate type I or III IFN immune responses. Zhang et al. identified Ku70 specifically as the DNA sensor and showed IRF1-IRF7 activation and expression of type III IFNs.149 Ferguson et al. determined the type I IFN response was independent of DNA-PK kinase activity, agreeing with the previously mentioned publication indicating Ku70 is the specific sensor. The authors were able to show DNA-PK contributing to the type I IFN immune response both in vitro and in vivo with a DNA-PK knockout mouse. They hypothesized DNA-PK signals through the STING-TBK1 pathway and showed activation of TBK1. Neither group adequately addressed the contribution of STING.150 However, recent work expanded upon the mechanism of Ku70 recognizing DNA in the cytosol and the requirement of STING-IRF1-IRF7 to stimulate a type III IFN response.151,152
These publications either showed or assumed a requirement of STING. Burleigh et al., however, revealed DNA-PK induction of type I IFNs that is both STING- and TBK1-independent. They discovered this pathway after still seeing IRF3 phosphorylation in STING or TBK1 knockout cells treated with calf thymus DNA. Contrary to the other publications, their results suggest that all three subunits of DNA-PK are required (Figure 3).153 It has been previously shown that DNA-PK can directly phosphorylate IRF3, but at different amino acid sites than those in Burleigh et al.154 More recently, another group found the Ku complex recognizes accumulated cytoplasmic DNA in aged human and mouse CD4+ T cells in a cGAS-STING independent manner, leading to T cell activation and an autoimmune pathology in aged mice. However, the data indicates a different mechanism than Burleigh et al. DNA-PK sensing of DNA leads to the phosphorylation of the ZAK kinase, which then activates AKT and the mTOR pathways.155
Taken altogether, the DNA-PK complex appears to be able to act as a cytosolic innate immune DNA sensor. Though, it is likely to be secondary to the cGAS-STING DNA sensing pathway. However, there are many discrepancies and contradictions in the published literature, such as the role of STING downstream of DNA-PK and which IFNs are produced. These differences could be due to species specificity, different cell types, or even different assays. These issues, though, are not unique to DNA-PK. Most of the other DDR factors discussed in this section face these same issues. Therefore, more work must be done to understand whether these differences are due to experimental set-up, choice of assays, data interpretation, or something more biologically important. Delving deeper will also help us understand how DDR factors switch from acting in the DDR to acting in innate immunity, and vice versa.
Perspective
Engagement and modulation of the DDR are central to the life cycles of a range of diverse viruses and are a phenotype that is broadly conserved beyond the viruses which we have discussed in this review. Despite the diversity in viral genomes, mechanisms of replication, and subcellular localization, the commonality of DDR engagement collectively highlights the importance of engaging ATM, ATR, and DNA-PK signaling pathways as well as the individual proteins in these pathways to promote the viral lifecycle. Converse to the benefits of utilizing the DDR, many of these factors themselves have antiviral activity as well as connections to innate immunity. As such, it is evolutionarily imperative for viruses to overcome this, and further demonstrates the convergence of these two important cellular functions which reflect two sides of the same coin that functions to sense, signal, and respond to aberrant nucleic acid – whether this be self or non-self.
Many questions remain as we are only just beginning to scratch the surface of the role the DDR plays in viral replication and innate immunity. For example, it is still unclear for many viruses whether steps such as induction of DNA damage or cell cycle arrest are active processes required for viral replication or consequences of other steps in viral replication. One way we propose to tackle this is to look for evolutionary conservation within viral genera. While conserved “byproducts” may exist, like spandrels in cathedrals,156 conservation of function is a strong indicator of significance in viral replication. It will also be important to directly address the causal relationships between the many ways a specific virus engages the DDR. Many steps in DDR signaling are intertwined, and most viruses we discussed activate and/or repress multiple aspects of the DDR. Thus, how one phenotype may influence another, such as how viruses induce transcriptional changes as a cellular consequence of DNA damage or whether there is a correlation between repression of DNA repair and changes in chromatin dynamics, will be essential to identify important DDR-associated drivers of viral replication. Moreover, determining what viral proteins overcome DDR proteins acting as innate immune proteins, and what additional DDR factors have important roles in innate immunity, is paramount to uncovering the connection between these two interconnected signaling pathways. One area of innate immunity where the DDR may be especially poised is in nuclear surveillance. It is clear from the literature that the cell contains a vast array of cytosolic innate immune sensors; however, nuclear innate immune sensors, such as cGAS, are only now just beginning to emerge. The DDR is perfectly situated to function in this role, and by systematically assessing potential roles of DDR proteins in both the nuclear and cytoplasmic innate immune response, we can further expand on these emerging fields. At the technical level, one aspect that must be addressed to help answer many of these questions is the use of consistent and reproducible assays that have been pioneered by the DDR field but are often overlooked by virologists.
We should consider how we can leverage the interconnection with the DDR to establish new therapeutics to treat viral infection. As the DDR is a major therapeutic target for cancer therapy, many drugs already exist that could be screened for antiviral roles, such as those found to inhibit SARS-CoV-2 replication.157 Viral dysregulation of the DDR could also be used to selectively deplete infected cells. For example, a direct outcome of DNA damage is that infected cells are hypersensitive to additional DNA damage. In both HTLV-1 and HIV infected cells, induction of DNA damage or repression of DNA repair makes infected cells hypersensitive to additional low levels of exogenous DSBs.10,15 Moreover, patients with HPV+ head and neck tumors have increased sensitivity and long-term survival when treated with chemotherapy that induces DNA damage.158 This concept of “synthetic lethality” has been proposed to treat many types of cancer that are deficient in DNA repair.159 For example, BRCA1/2-deficient tumors are highly susceptible to inhibition of the DNA repair protein PARP1 due to the inability to repair double-strand breaks by HR or NHEJ.160,161 Given the ability of diverse viruses to induce DNA damage and/or antagonize DDR signaling and repair, we propose that a synthetic lethality approach may be feasible to selectively deplete infected cells. Based on the breadth of drugs available to induce low levels of genotoxic stress (including orphaned drugs and others that never made it to clinical trials), it will be important to thoroughly assess the efficacy of a synthetic lethality approach to killing infected cells in vitro and in vivo.
Acknowledgments
This work was supported by NIH NIAID R01AI147837 to O.I.F. A.L., R.N.D, and V.Y. have been supported by the Ruth L. Kirschstein National Research Service Award “Molecular Pathogenesis Training Grant” AI007323. C. S. has been supported by the Ruth L. Kirschstein National Research Service Award “Cell and Molecular Biology Training Grant” GM007185. A.L. and C.S. have been supported by the Eugene V. Cota-Robles Fellowship. We thank BioRender for figures created on BioRender.com. We sincerely thank Dr. Patrick S. Mitchell (University of Washington) for his comments on the manuscript.
Abbreviations:
- Ad
adenovirus
- ATR
ataxia telangiectasia and Rad3related protein
- ATM
ataxia-telangiectasia mutated
- cGAS
cyclic GMP–AMP synthase
- DDR
DNA damage response
- DNA-PK
Ku70, Ku80, and DNA-PKcs
- DSB
double-stranded DNA break
- dsDNA
double-stranded DNA
- E
early
- EBV
Epstein-Barr virus
- HSV
herpes simplex virus
- HR
homologous recombination
- HEK
human embryonic kidney
- HIV
human immunodeficiency virus
- HPV
human papillomavirus
- HTLV
human T-lymphotropic virus
- IBV
infectious bronchitis virus
- IAV
influenza A
- IFI16
nterferon gamma inducible protein 16
- IRF
interferon regulatory factor
- IFN
interferon
- KSHV
Kaposi’s sarcoma-associated herpesvirus
- LANA
latency-associated nuclear antigen
- MMR
mismatch repair
- MRE11 meiotic recombination 11
MRN, MRE11, Rad50, NBS1
- NEHJ
nonhomologous end joining
- NF-κB
nuclear factor kappa B
- PARP1
poly(ADP) ribose polymerase
- PS
positive selection
- PTM
post-translational modification
- PFGE
pulsed-field gel electrophoresis
- RPA
replication protein A
- SAMHD1
SAM domain and HD domain-containing protein 1
- SV40
simian virus 40
- SSB
single-strand DNA break
- ssDNA
single-stranded DNA
- STING
stimulator of interferon genes
Footnotes
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
CRediT authorship contribution statement
Andrew Lopez: Conceptualization, Writing – original draft, Visualization. Randilea Nichols Doyle: Conceptualization, Writing – original draft. Carina Sandoval: Conceptualization, Writing – original draft. Karly Nisson: Writing – original draft. Vivian Yang: Writing – original draft. Oliver I. Fregoso: Conceptualization, Writing – review & editing.
References
- 1.Baltimore D, (1971). Expression of animal virus genomes. Bacteriol. Rev. 35, 235–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Weitzman MD, Fradet-Turcotte A, (2018). Virus DNA Replication and the Host DNA Damage Response. Annu. Rev. Virol. 5, 141–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Luftig MA, (2014). Viruses and the DNA Damage Response: Activation and Antagonism. Annu. Rev. Virol. 1, 605–625. [DOI] [PubMed] [Google Scholar]
- 4.Turnell AS, Grand RJ, (2012). DNA viruses and the cellular DNA-damage response. J. Gen. Virol. 93, 2076–2097. [DOI] [PubMed] [Google Scholar]
- 5.Blackford AN, Jackson SP, (2017). ATM, ATR, and DNA-PK: The Trinity at the Heart of the DNA Damage Response. Mol. Cell 66, 801–817. [DOI] [PubMed] [Google Scholar]
- 6.Ciccia A, Elledge SJ, (2010). The DNA damage response: making it safe to play with knives. Mol. Cell 40, 179–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Boichuk S, Hu L, Hein J, Gjoerup OV, (2010). Multiple DNA damage signaling and repair pathways deregulated by simian virus 40 large T antigen. J. Virol. 84, 8007–8020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zur HH, (1967). Induction of specific chromosomal aberrations by adenovirus type 12 in human embryonic kidney cells. J. Virol. 1, 1174–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Vijaya Lakshmi AN, Ramana MV, Vijayashree B, Ahuja YR, Sharma G, (1999). Detection of influenza virus induced DNA damage by comet assay. Mutat. Res. 442, 53–58. [DOI] [PubMed] [Google Scholar]
- 10.Baydoun HH, Bai XT, Shelton S, Nicot C, (2012). HTLV-I tax increases genetic instability by inducing DNA double strand breaks during DNA replication and switching repair to NHEJ. PLoS ONE 7, e42226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Skalka AM, Katz RA, (2005). Retroviral DNA integration and the DNA damage response. Cell Death Differ. 12 (Suppl 1), 971–978. [DOI] [PubMed] [Google Scholar]
- 12.Koyama T, Sun B, Tokunaga K, Tatsumi M, Ishizaka Y, (2013). DNA damage enhances integration of HIV-1 into macrophages by overcoming integrase inhibition. Retrovirology 10, 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Iijima K, Kobayashi J, Ishizaka Y, (2018). Structural alteration of DNA induced by viral protein R of HIV-1 triggers the DNA damage response. Retrovirology 15, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tachiwana H, Shimura M, Nakai-Murakami C, Tokunaga K, Takizawa Y, Sata T, et al. , (2006). HIV-1 Vpr induces DNA double-strand breaks. Cancer Res. 66, 627–631. [DOI] [PubMed] [Google Scholar]
- 15.Li D, Lopez A, Sandoval C, Nichols Doyle R, Fregoso OI, (2020). HIV Vpr Modulates the Host DNA Damage Response at Two Independent Steps to Damage DNA and Repress Double-Strand DNA Break Repair. mBio 11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhang S, Pointer D, Singer G, Feng Y, Park K, Zhao LJ, (1998). Direct binding to nucleic acids by Vpr of human immunodeficiency virus type 1. Gene 212, 157–166. [DOI] [PubMed] [Google Scholar]
- 17.Lai M, Zimmerman ES, Planelles V, Chen J, (2005). Activation of the ATR pathway by human immunodeficiency virus type 1 Vpr involves its direct binding to chromatin in vivo. J. Virol. 79, 15443–15451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Murugesapillai D, Bouaziz S, Maher LJ, Israeloff NE, Cameron CE, Williams MC, (2017). Accurate nanoscale flexibility measurement of DNA and DNA-protein complexes by atomic force microscopy in liquid. Nanoscale 9, 11327–11337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zimmerman ES, Sherman MP, Blackett JL, Neidleman JA, Kreis C, Mundt P, et al. , (2006). Human immunodeficiency virus type 1 Vpr induces DNA replication stress in vitro and in vivo. J. Virol. 80, 10407–10418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Schrofelbauer B, Yu Q, Zeitlin SG, Landau NR, (2005). Human immunodeficiency virus type 1 Vpr induces the degradation of the UNG and SMUG uracil-DNA glycosylases. J. Virol. 79, 10978–10987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wu Y, Zhou X, Barnes CO, DeLucia M, Cohen AE, Gronenborn AM, et al. , (2016). The DDB1-DCAF1-Vpr-UNG2 crystal structure reveals how HIV-1 Vpr steers human UNG2 toward destruction. Nature Struct. Mol. Biol. 23, 933–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hrecka K, Hao C, Shun MC, Kaur S, Swanson SK, Florens L, et al. , (2016). HIV-1 and HIV-2 exhibit divergent interactions with HLTF and UNG2 DNA repair proteins. Proc. Natl. Acad. Sci. U. S. A. 113, E3921–E3930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lahouassa H, Blondot ML, Chauveau L, Chougui G, Morel M, Leduc M, et al. , (2016). HIV-1 Vpr degrades the HLTF DNA translocase in T cells and macrophages. Proc. Natl. Acad. Sci. U. S. A. 113, 5311–5316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Laguette N, Bregnard C, Hue P, Basbous J, Yatim A, Larroque M, et al. , (2014). Premature activation of the SLX4 complex by Vpr promotes G2/M arrest and escape from innate immune sensing. Cell 156, 134–145. [DOI] [PubMed] [Google Scholar]
- 25.Zhou X, DeLucia M, Ahn J, (2016). SLX4-SLX1 Protein-independent Down-regulation of MUS81-EME1 Protein by HIV-1 Viral Protein R (Vpr). J. Biol. Chem. 291, 16936–16947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yan J, Shun MC, Hao C, Zhang Y, Qian J, Hrecka K, et al. , (2018). HIV-1 Vpr Reprograms CLR4(DCAF1) E3 Ubiquitin Ligase to Antagonize Exonuclease 1-Mediated Restriction of HIV-1 Infection. mBio 9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lv L, Wang Q, Xu Y, Tsao LC, Nakagawa T, Guo H, et al. , (2018). Vpr Targets TET2 for Degradation by CRL4(VprBP) E3 Ligase to Sustain IL-6 Expression and Enhance HIV-1 Replication. Mol. Cell 70 961–70 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Romani B, Shaykh Baygloo N, Aghasadeghi MR, Allahbakhshi E, (2015). HIV-1 Vpr Protein Enhances Proteasomal Degradation of MCM10 DNA Replication Factor through the Cul4-DDB1[VprBP] E3 Ubiquitin Ligase to Induce G2/M Cell Cycle Arrest. J. Biol. Chem. 290, 17380–17389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Withers-Ward ES, Jowett JB, Stewart SA, Xie YM, Garfinkel A, Shibagaki Y, et al. , (1997). Human immunodeficiency virus type 1 Vpr interacts with HHR23A, a cellular protein implicated in nucleotide excision DNA repair. J. Virol. 71, 9732–9742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lim ES, Fregoso OI, McCoy CO, Matsen FA, Malik HS, Emerman M, (2012). The ability of primate lentiviruses to degrade the monocyte restriction factor SAMHD1 preceded the birth of the viral accessory protein Vpx. Cell Host Microbe 11, 194–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fregoso OI, Ahn J, Wang C, Mehrens J, Skowronski J, Emerman M, (2013). Evolutionary toggling of Vpx/Vpr specificity results in divergent recognition of the restriction factor SAMHD1. PLoS Pathog. 9, e1003496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fabryova H, Strebel K, (2019). Vpr and Its Cellular Interaction Partners: R We There Yet? Cells 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Greenwood EJD, Williamson JC, Sienkiewicz A, Naamati A, Matheson NJ, Lehner PJ, (2019). Promiscuous Targeting of Cellular Proteins by Vpr Drives Systems-Level Proteomic Remodeling in HIV-1 Infection. Cell Rep 27 1579–96e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lentz TB, Samulski RJ, (2015). Insight into the mechanism of inhibition of adeno-associated virus by the Mre11/Rad50/Nbs1 complex. J. Virol. 89, 181–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lakdawala SS, Schwartz RA, Ferenchak K, Carson CT, McSharry BP, Wilkinson GW, et al. , (2008). Differential requirements of the C terminus of Nbs1 in suppressing adenovirus DNA replication and promoting concatemer formation. J. Virol. 82, 8362–8372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sarkar R, Patra U, Lo M, Mukherjee A, Biswas A, Chawla-Sarkar M, (2020). Rotavirus activates a noncanonical ATM-Chk2 branch of DNA damage response during infection to positively regulate viroplasm dynamics. Cell. Microbiol. 22, e13149. [DOI] [PubMed] [Google Scholar]
- 37.Evans JD, Hearing P, (2005). Relocalization of the Mre11-Rad50-Nbs1 complex by the adenovirus E4 ORF3 protein is required for viral replication. J. Virol. 79, 6207–6215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Liu Y, Shevchenko A, Shevchenko A, Berk AJ, (2005). Adenovirus exploits the cellular aggresome response to accelerate inactivation of the MRN complex. J. Virol. 79, 14004–14016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Forrester NA, Sedgwick GG, Thomas A, Blackford AN, Speiseder T, Dobner T, et al. , (2011). Serotype-specific inactivation of the cellular DNA damage response during adenovirus infection. J. Virol. 85, 2201–2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cheng CY, Gilson T, Dallaire F, Ketner G, Branton PE, Blanchette P, (2011). The E4orf6/E1B55K E3 ubiquitin ligase complexes of human adenoviruses exhibit heterogeneity in composition and substrate specificity. J. Virol. 85, 765–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pancholi NJ, Weitzman MD, (2018). Serotype-specific restriction of wild-type adenoviruses by the cellular Mre11-Rad50-Nbs1 complex. Virology 518, 221–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mariggio G, Koch S, Zhang G, Weidner-Glunde M, Ruckert J, Kati S, et al. , (2017). Kaposi Sarcoma Herpesvirus (KSHV) Latency-Associated Nuclear Antigen (LANA) recruits components of the MRN (Mre11-Rad50-NBS1) repair complex to modulate an innate immune signaling pathway and viral latency. PLoS Pathog. 13, e1006335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Baydoun HH, Pancewicz J, Nicot C, (2011). Human T-lymphotropic type 1 virus p30 inhibits homologous recombination and favors unfaithful DNA repair. Blood 117, 5897–5906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Blackford AN, Bruton RK, Dirlik O, Stewart GS, Taylor AM, Dobner T, et al. , (2008). A role for E1B-AP5 in ATR signaling pathways during adenovirus infection. J. Virol. 82, 7640–7652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ducu RI, Dayaram T, Marriott SJ, (2011). The HTLV-1 Tax oncoprotein represses Ku80 gene expression. Virology 416, 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wang J, Kang L, Song D, Liu L, Yang S, Ma L, et al. , (2017). Ku70 Senses HTLV-1 DNA and Modulates HTLV-1 Replication. J. Immunol. 199, 2475–2482. [DOI] [PubMed] [Google Scholar]
- 47.Burkhart DL, Sage J, (2008). Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nature Rev. Cancer 8, 671–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Joerger AC, Fersht AR, (2016). The p53 Pathway: Origins, Inactivation in Cancer, and Emerging Therapeutic Approaches. Annu. Rev. Biochem. 85, 375–404. [DOI] [PubMed] [Google Scholar]
- 49.Bieging KT, Mello SS, Attardi LD, (2014). Unravelling mechanisms of p53-mediated tumour suppression. Nature Rev. Cancer 14, 359–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Dyson N, Howley PM, Munger K, Harlow E, (1989). The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 243, 934–937. [DOI] [PubMed] [Google Scholar]
- 51.Munger K, Phelps WC, Bubb V, Howley PM, Schlegel R, (1989). The E6 and E7 genes of the human papillomavirus type 16 together are necessary and sufficient for transformation of primary human keratinocytes. J. Virol. 63, 4417–4421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Moody CA, Laimins LA, (2009). Human papillomaviruses activate the ATM DNA damage pathway for viral genome amplification upon differentiation. PLoS Pathog. 5, e1000605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Baker A, Rohleder KJ, Hanakahi LA, Ketner G, (2007). Adenovirus E4 34k and E1b 55k oncoproteins target host DNA ligase IV for proteasomal degradation. J. Virol. 81, 7034–7040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Boyer J, Rohleder K, Ketner G, (1999). Adenovirus E4 34k and E4 11k inhibit double strand break repair and are physically associated with the cellular DNA-dependent protein kinase. Virology 263, 307–312. [DOI] [PubMed] [Google Scholar]
- 55.Gluck S, Buttafuoco A, Meier AF, Arnoldi F, Vogt B, Schraner EM, et al. , (2017). Rotavirus replication is correlated with S/G2 interphase arrest of the host cell cycle. PLoS ONE 12, e0179607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Jiang W, Wang Q, Chen S, Gao S, Song L, Liu P, et al. , (2013). Influenza A virus NS1 induces G0/G1 cell cycle arrest by inhibiting the expression and activity of RhoA protein. J. Virol. 87, 3039–3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Li N, Parrish M, Chan TK, Yin L, Rai P, Yoshiyuki Y, et al. , (2015). Influenza infection induces host DNA damage and dynamic DNA damage responses during tissue regeneration. Cell. Mol. Life Sci. 72, 2973–2988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wallace NA, Khanal S, Robinson KL, Wendel SO, Messer JJ, Galloway DA, (2017). High-Risk Alphapapillomavirus Oncogenes Impair the Homologous Recombination Pathway. J. Virol. 91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mehta K, Laimins L, (2018). Human Papillomaviruses Preferentially Recruit DNA Repair Factors to Viral Genomes for Rapid Repair and Amplification. mBio 9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Park JW, Nickel KP, Torres AD, Lee D, Lambert PF, Kimple RJ, (2014). Human papillomavirus type 16 E7 oncoprotein causes a delay in repair of DNA damage. Radiother. Oncol. 113, 337–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Nakai-Murakami C, Shimura M, Kinomoto M, Takizawa Y, Tokunaga K, Taguchi T, et al. , (2007). HIV-1 Vpr induces ATM-dependent cellular signal with enhanced homologous recombination. Oncogene 26, 477–486. [DOI] [PubMed] [Google Scholar]
- 62.Chambers BS, Heaton BE, Rausch K, Dumm RE, Hamilton JR, Cherry S, et al. , (2019). DNA mismatch repair is required for the host innate response and controls cellular fate after influenza virus infection. Nature Microbiol. 4, 1964–1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sato Y, Tsurumi T, (2010). Noise cancellation: viral fine tuning of the cellular environment for its own genome replication. PLoS Pathog. 6, e1001158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bagga S, Bouchard MJ, (2014). Cell cycle regulation during viral infection. Methods Mol. Biol. 1170, 165–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ben-Israel H, Kleinberger T, (2002). Adenovirus and cell cycle control. Front. Biosci. 7, d1369–d1395. [DOI] [PubMed] [Google Scholar]
- 66.McFadden K, Luftig MA, (2013). Interplay between DNA tumor viruses and the host DNA damage response. Curr. Top. Microbiol. Immunol. 371, 229–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Fan Y, Sanyal S, Bruzzone R, (2018). Breaking Bad: How Viruses Subvert the Cell Cycle. Front. Cell. Infect. Microbiol. 8, 396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ahuja D, Saenz-Robles MT, Pipas JM, (2005). SV40 large T antigen targets multiple cellular pathways to elicit cellular transformation. Oncogene 24, 7729–7745. [DOI] [PubMed] [Google Scholar]
- 69.Yuan X, Yao Z, Wu J, Zhou Y, Shan Y, Dong B, et al. , (2007). G1 phase cell cycle arrest induced by SARS-CoV 3a protein via the cyclin D3/pRb pathway. Am. J. Respir. Cell Mol. Biol. 37, 9–19. [DOI] [PubMed] [Google Scholar]
- 70.Yuan X, Wu J, Shan Y, Yao Z, Dong B, Chen B, et al. , (2006). SARS coronavirus 7a protein blocks cell cycle progression at G0/G1 phase via the cyclin D3/pRb pathway. Virology 346, 74–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Surjit M, Liu B, Chow VT, Lal SK, (2006). The nucleocapsid protein of severe acute respiratory syndrome-coronavirus inhibits the activity of cyclin-cyclin-dependent kinase complex and blocks S phase progression in mammalian cells. J. Biol. Chem. 281, 10669–10681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Li FQ, Tam JP, Liu DX, (2007). Cell cycle arrest and apoptosis induced by the coronavirus infectious bronchitis virus in the absence of p53. Virology 365, 435–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Dayaram T, Lemoine FJ, Donehower LA, Marriott SJ, (2013). Activation of WIP1 phosphatase by HTLV-1 Tax mitigates the cellular response to DNA damage. PLoS ONE 8, e55989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Haoudi A, Daniels RC, Wong E, Kupfer G, Semmes OJ, (2003). Human T-cell leukemia virus-I tax oncoprotein functionally targets a subnuclear complex involved in cellular DNA damage-response. J. Biol. Chem. 278, 37736–37744. [DOI] [PubMed] [Google Scholar]
- 75.Gupta SK, Guo X, Durkin SS, Fryrear KF, Ward MD, Semmes OJ, (2007). Human T-cell leukemia virus type 1 Tax oncoprotein prevents DNA damage-induced chromatin egress of hyperphosphorylated Chk2. J. Biol. Chem. 282, 29431–29440. [DOI] [PubMed] [Google Scholar]
- 76.Salamango DJ, Ikeda T, Moghadasi SA, Wang J, McCann JL, Serebrenik AA, et al. , (2019). HIV-1 Vif Triggers Cell Cycle Arrest by Degrading Cellular PPP2R5 Phospho-regulators. Cell Rep 29 1057–65 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Stivahtis GL, Soares MA, Vodicka MA, Hahn BH, Emerman M, (1997). Conservation and host specificity of Vpr-mediated cell cycle arrest suggest a fundamental role in primate lentivirus evolution and biology. J. Virol. 71, 4331–4338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Planelles V, Jowett JB, Li QX, Xie Y, Hahn B, Chen IS, (1996). Vpr-induced cell cycle arrest is conserved among primate lentiviruses. J. Virol. 70, 2516–2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Clark E, Santiago F, Deng L, Chong S, de La Fuente C, Wang L, et al. , (2000). Loss of G(1)/S checkpoint in human immunodeficiency virus type 1-infected cells is associated with a lack of cyclin-dependent kinase inhibitor p21/Waf1. J. Virol. 74, 5040–5052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Goh WC, Rogel ME, Kinsey CM, Michael SF, Fultz PN, Nowak MA, et al. , (1998). HIV-1 Vpr increases viral expression by manipulation of the cell cycle: a mechanism for selection of Vpr in vivo. Nature Med. 4, 65–71. [DOI] [PubMed] [Google Scholar]
- 81.Miller CM, Akiyama H, Agosto LM, Emery A, Ettinger CR, Swanstrom RI, et al. , (2017). Virion-Associated Vpr Alleviates a Postintegration Block to HIV-1 Infection of Dendritic Cells. J. Virol. 91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Nakada S, (2016). Opposing roles of RNF8/RNF168 and deubiquitinating enzymes in ubiquitination-dependent DNA double-strand break response signaling and DNA-repair pathway choice. J Radiat Res. 57 (Suppl 1), i33–i40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lilley CE, Chaurushiya MS, Boutell C, Landry S, Suh J, Panier S, et al. , (2010). A viral E3 ligase targets RNF8 and RNF168 to control histone ubiquitination and DNA damage responses. EMBO J. 29, 943–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Sitz J, Blanchet SA, Gameiro SF, Biquand E, Morgan TM, Galloy M, et al. , (2019). Human papillomavirus E7 oncoprotein targets RNF168 to hijack the host DNA damage response. Proc. Natl. Acad. Sci. U. S. A. 116, 19552–19562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Yang J, Deng W, Hau PM, Liu J, Lau VM, Cheung AL, et al. , (2015). Epstein-Barr virus BZLF1 protein impairs accumulation of host DNA damage proteins at damage sites in response to DNA damage. Lab. Invest. 95, 937–950. [DOI] [PubMed] [Google Scholar]
- 86.Ho YK, Zhi H, Bowlin T, Dorjbal B, Philip S, Zahoor MA, et al. , (2015). HTLV-1 Tax Stimulates Ubiquitin E3 Ligase, Ring Finger Protein 8, to Assemble Lysine 63-Linked Polyubiquitin Chains for TAK1 and IKK Activation. PLoS Pathog. 11, e1005102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Squatrito M, Gorrini C, Amati B, (2006). Tip60 in DNA damage response and growth control: many tricks in one HAT. Trends Cell Biol. 16, 433–442. [DOI] [PubMed] [Google Scholar]
- 88.Sun Y, Jiang X, Chen S, Fernandes N, Price BD, (2005). A role for the Tip60 histone acetyltransferase in the acetylation and activation of ATM. Proc. Natl. Acad. Sci. U. S. A. 102, 13182–13187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kamine J, Elangovan B, Subramanian T, Coleman D, Chinnadurai G, (1996). Identification of a cellular protein that specifically interacts with the essential cysteine region of the HIV-1 Tat transactivator. Virology 216, 357–366. [DOI] [PubMed] [Google Scholar]
- 90.Creaven M, Hans F, Mutskov V, Col E, Caron C, Dimitrov S, et al. , (1999). Control of the histone-acetyltransferase activity of Tip60 by the HIV-1 transactivator protein. Tat. Biochem. 38, 8826–8830. [DOI] [PubMed] [Google Scholar]
- 91.Col E, Caron C, Chable-Bessia C, Legube G, Gazzeri S, Komatsu Y, et al. , (2005). HIV-1 Tat targets Tip60 to impair the apoptotic cell response to genotoxic stresses. EMBO J. 24, 2634–2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zhang SM, Song M, Yang TY, Fan R, Liu XD, Zhou PK, (2012). HIV-1 Tat impairs cell cycle control by targeting the Tip60, Plk1 and cyclin B1 ternary complex. Cell Cycle 11, 1217–1234. [DOI] [PubMed] [Google Scholar]
- 93.Awasthi S, Sharma A, Wong K, Zhang J, Matlock EF, Rogers L, et al. , (2005). A human T-cell lymphotropic virus type 1 enhancer of Myc transforming potential stabilizes Myc-TIP60 transcriptional interactions. Mol. Cell. Biol. 25, 6178–6198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gupta A, Jha S, Engel DA, Ornelles DA, Dutta A, (2013). Tip60 degradation by adenovirus relieves transcriptional repression of viral transcriptional activator EIA. Oncogene 32, 5017–5025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Li R, Zhu J, Xie Z, Liao G, Liu J, Chen MR, et al. , (2011). Conserved herpesvirus kinases target the DNA damage response pathway and TIP60 histone acetyltransferase to promote virus replication. Cell Host Microbe 10, 390–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Reitsma JM, Savaryn JP, Faust K, Sato H, Halligan BD, Terhune SS, (2011). Antiviral inhibition targeting the HCMV kinase pUL97 requires pUL27-dependent degradation of Tip60 acetyltransferase and cell-cycle arrest. Cell Host Microbe 9, 103–114. [DOI] [PubMed] [Google Scholar]
- 97.Shamay M, Liu J, Li R, Liao G, Shen L, Greenway M, et al. , (2012). A protein array screen for Kaposi’s sarcoma-associated herpesvirus LANA interactors links LANA to TIP60, PP2A activity, and telomere shortening. J. Virol. 86, 5179–5191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Jha S, Vande Pol S, Banerjee NS, Dutta AB, Chow LT, Dutta A, (2010). Destabilization of TIP60 by human papillomavirus E6 results in attenuation of TIP60-dependent transcriptional regulation and apoptotic pathway. Mol. Cell 38, 700–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Subbaiah VK, Zhang Y, Rajagopalan D, Abdullah LN, Yeo-Teh NS, Tomaic V, et al. , (2016). E3 ligase EDD1/UBR5 is utilized by the HPV E6 oncogene to destabilize tumor suppressor TIP60. Oncogene 35, 2062–2074. [DOI] [PubMed] [Google Scholar]
- 100.Hong S, Dutta A, Laimins LA, (2015). The acetyltransferase Tip60 is a critical regulator of the differentiation-dependent amplification of human papillomaviruses. J. Virol. 89, 4668–4675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Smith JA, Haberstroh FS, White EA, Livingston DM, DeCaprio JA, Howley PM, (2014). SMCX and components of the TIP60 complex contribute to E2 regulation of the HPV E6/E7 promoter. Virology 468–470, 311–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Nishitsuji H, Ujino S, Harada K, Shimotohno K, (2018). TIP60 Complex Inhibits Hepatitis B Virus Transcription. J. Virol. 92 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ma G, Chen L, Luo J, Wang B, Wang C, Li M, et al. , (2018). Histone acetyl transferase TIP60 inhibits the replication of influenza a virus by activation the TBK1-IRF3 pathway. Virol J. 15, 172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Heine GF, Horwitz AA, Parvin JD, (2008). Multiple mechanisms contribute to inhibit transcription in response to DNA damage. J. Biol. Chem. 283, 9555–9561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Huang TT, Wuerzberger-Davis SM, Wu ZH, Miyamoto S, (2003). Sequential modification of NEMO/IKKgamma by SUMO-1 and ubiquitin mediates NF-kappaB activation by genotoxic stress. Cell 115, 565–576. [DOI] [PubMed] [Google Scholar]
- 106.Medunjanin S, Putzier M, Nothen T, Weinert S, Kahne T, Luani B, et al. , (2020). DNA-PK: gatekeeper for IKKgamma/NEMO nucleocytoplasmic shuttling in genotoxic stress-induced NF-kappaB activation. Cell. Mol. Life Sci. 77, 4133–4142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Rashi-Elkeles S, Warnatz HJ, Elkon R, Kupershtein A, Chobod Y, Paz A, et al. , (2014). Parallel profiling of the transcriptome, cistrome, and epigenome in the cellular response to ionizing radiation. Sci Signal. 7, rs3. [DOI] [PubMed] [Google Scholar]
- 108.McCool KW, Miyamoto S, (2012). DNA damage-dependent NF-kappaB activation: NEMO turns nuclear signaling inside out. Immunol. Rev. 246, 311–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Millen S, Meretuk L, Gottlicher T, Schmitt S, Fleckenstein B, Thoma-Kress AK, (2020). A novel positive feedback-loop between the HTLV-1 oncoprotein Tax and NF-kappaB activity in T-cells. Retrovirology 17, 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kinjo T, Ham-Terhune J, Peloponese JM Jr., Jeang KT, (2010). Induction of reactive oxygen species by human T-cell leukemia virus type 1 tax correlates with DNA damage and expression of cellular senescence marker. J. Virol. 84, 5431–5437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Mitchell S, Vargas J, Hoffmann A, (2016). Signaling via the NFkappaB system. Wiley Interdiscip. Rev. Syst. Biol. Med. 8, 227–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Heusinger E, Kirchhoff F, (2017). Primate Lentiviruses Modulate NF-kappaB Activity by Multiple Mechanisms to Fine-Tune Viral and Cellular Gene Expression. Front. Microbiol. 8, 198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Nakahara T, Tanaka K, Ohno S, Egawa N, Yugawa T, Kiyono T, (2015). Activation of NF-kappaB by human papillomavirus 16 E1 limits E1-dependent viral replication through degradation of E1. J. Virol. 89, 5040–5059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Irwan ID, Cullen BR, (2021). Tax Induces the Recruitment of NF-kappaB to Unintegrated HIV-1 DNA To Rescue Viral Gene Expression and Replication. J. Virol. 95, e0028521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Harhaj EW, Giam CZ, (2018). NF-kappaB signaling mechanisms in HTLV-1-induced adult T-cell leukemia/lymphoma. FEBS J. 285, 3324–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Valerie K, Delers A, Bruck C, Thiriart C, Rosenberg H, Debouck C, et al. , (1988). Activation of human immunodeficiency virus type 1 by DNA damage in human cells. Nature 333, 78–81. [DOI] [PubMed] [Google Scholar]
- 117.Zierhut C, Yamaguchi N, Paredes M, Luo JD, Carroll T, Funabiki H, (2019). The Cytoplasmic DNA Sensor cGAS Promotes Mitotic Cell Death. Cell 178 302–15 e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Mackenzie KJ, Carroll P, Martin CA, Murina O, Fluteau A, Simpson DJ, et al. , (2017). cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548, 461–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Greenberg RA, (2017). Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 548, 466–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Liu H, Zhang H, Wu X, Ma D, Wu J, Wang L, et al. , (2018). Nuclear cGAS suppresses DNA repair and promotes tumorigenesis. Nature 563, 131–136. [DOI] [PubMed] [Google Scholar]
- 121.Jiang H, Xue X, Panda S, Kawale A, Hooy RM, Liang F, et al. , (2019). Chromatin-bound cGAS is an inhibitor of DNA repair and hence accelerates genome destabilization and cell death. EMBO J. 38, e102718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ma Z, Damania B, (2016). The cGAS-STING Defense Pathway and Its Counteraction by Viruses. Cell Host Microbe 19, 150–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Takeuchi O, Akira S, (2009). Innate immunity to virus infection. Immunol. Rev. 227, 75–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Laguette N, Sobhian B, Casartelli N, Ringeard M, Chable-Bessia C, Segeral E, et al. , (2011). SAMHD1 is the dendritic- and myeloid-cell-specific HIV-1 restriction factor counteracted by Vpx. Nature 474, 654–657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hrecka K, Hao C, Gierszewska M, Swanson SK, Kesik-Brodacka M, Srivastava S, et al. , (2011). Vpx relieves inhibition of HIV-1 infection of macrophages mediated by the SAMHD1 protein. Nature 474, 658–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Laguette N, Rahm N, Sobhian B, Chable-Bessia C, Munch J, Snoeck J, et al. , (2012). Evolutionary and functional analyses of the interaction between the myeloid restriction factor SAMHD1 and the lentiviral Vpx protein. Cell Host Microbe 11, 205–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kretschmer S, Wolf C, Konig N, Staroske W, Guck J, Hausler M, et al. , (2015). SAMHD1 prevents autoimmunity by maintaining genome stability. Ann. Rheum. Dis. 74, e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Clifford R, Louis T, Robbe P, Ackroyd S, Burns A, Timbs AT, et al. , (2014). SAMHD1 is mutated recurrently in chronic lymphocytic leukemia and is involved in response to DNA damage. Blood 123, 1021–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zeman MK, Cimprich KA, (2014). Causes and consequences of replication stress. Nature Cell Biol. 16, 2–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Daddacha W, Koyen AE, Bastien AJ, Head PE, Dhere VR, Nabeta GN, et al. , (2017). SAMHD1 Promotes DNA End Resection to Facilitate DNA Repair by Homologous Recombination. Cell Rep 20, 1921–1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Coquel F, Silva MJ, Techer H, Zadorozhny K, Sharma S, Nieminuszczy J, et al. , (2018). SAMHD1 acts at stalled replication forks to prevent interferon induction. Nature 557, 57–61. [DOI] [PubMed] [Google Scholar]
- 132.Mlcochova P, Caswell SJ, Taylor IA, Towers GJ, Gupta RK, (2018). DNA damage induced by topoisomerase inhibitors activates SAMHD1 and blocks HIV-1 infection of macrophages. EMBO J. 37, 50–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Jauregui P, Landau NR, (2018). DNA damage induces a SAMHD1-mediated block to the infection of macrophages by HIV-1. Sci. Rep. 8, 4153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Hadian K, Krappmann D, (2011). Signals from the nucleus: activation of NF-kappaB by cytosolic ATM in the DNA damage response. Sci Signal. 4, pe2. [DOI] [PubMed] [Google Scholar]
- 135.Hinz M, Stilmann M, Arslan SC, Khanna KK, Dittmar G, Scheidereit C, (2010). A cytoplasmic ATM-TRAF6-cIAP1 module links nuclear DNA damage signaling to ubiquitin-mediated NF-kappaB activation. Mol. Cell 40, 63–74. [DOI] [PubMed] [Google Scholar]
- 136.Wu ZH, Wong ET, Shi Y, Niu J, Chen Z, Miyamoto S, et al. , (2010). ATM- and NEMO-dependent ELKS ubiquitination coordinates TAK1-mediated IKK activation in response to genotoxic stress. Mol. Cell 40, 75–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Purbey PK, Scumpia PO, Kim PJ, Tong AJ, Iwamoto KS, McBride WH, et al. , (2017). Defined Sensing Mechanisms and Signaling Pathways Contribute to the Global Inflammatory Gene Expression Output Elicited by Ionizing Radiation. Immunity 47 421–34 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Dunphy G, Flannery SM, Almine JF, Connolly DJ, Paulus C, Jonsson KL, et al. , (2018). Non-canonical Activation of the DNA Sensing Adaptor STING by ATM and IFI16 Mediates NF-kappaB Signaling after Nuclear DNA Damage. Mol. Cell 71 745–60 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Hartlova A, Erttmann SF, Raffi FA, Schmalz AM, Resch U, Anugula S, et al. , (2015). DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 42, 332–343. [DOI] [PubMed] [Google Scholar]
- 140.Erttmann SF, Hartlova A, Sloniecka M, Raffi FA, Hosseinzadeh A, Edgren T, et al. , (2016). Loss of the DNA Damage Repair Kinase ATM Impairs Inflammasome-Dependent Anti-Bacterial Innate Immunity. Immunity 45, 106–118. [DOI] [PubMed] [Google Scholar]
- 141.Morales AJ, Carrero JA, Hung PJ, Tubbs AT, Andrews JM, Edelson BT, et al. , (2017). A type I IFN-dependent DNA damage response regulates the genetic program and inflammasome activation in macrophages. Elife 6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Kondo T, Kobayashi J, Saitoh T, Maruyama K, Ishii KJ, Barber GN, et al. , (2013). DNA damage sensor MRE11 recognizes cytosolic double-stranded DNA and induces type I interferon by regulating STING trafficking. Proc. Natl. Acad. Sci. U. S. A. 110, 2969–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Fu H, Martin MM, Regairaz M, Huang L, You Y, Lin CM, et al. , (2015). The DNA repair endonuclease Mus81 facilitates fast DNA replication in the absence of exogenous damage. Nature Commun 6, 6746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Lai X, Broderick R, Bergoglio V, Zimmer J, Badie S, Niedzwiedz W, et al. , (2017). MUS81 nuclease activity is essential for replication stress tolerance and chromosome segregation in BRCA2-deficient cells. Nature Commun. 8, 15983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ho SS, Zhang WY, Tan NY, Khatoo M, Suter MA, Tripathi S, et al. , (2016). The DNA Structure-Specific Endonuclease MUS81 Mediates DNA Sensor STING-Dependent Host Rejection of Prostate Cancer Cells. Immunity 44, 1177–1189. [DOI] [PubMed] [Google Scholar]
- 146.Ng KW, Marshall EA, Bell JC, Lam WL, (2018). cGAS-STING and Cancer: Dichotomous Roles in Tumor Immunity and Development. Trends Immunol. 39, 44–54. [DOI] [PubMed] [Google Scholar]
- 147.Wolf C, Rapp A, Berndt N, Staroske W, Schuster M, Dobrick-Mattheuer M, et al. , (2016). RPA and Rad51 constitute a cell intrinsic mechanism to protect the cytosol from self DNA. Nature Commun. 7, 11752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bhattacharya S, Srinivasan K, Abdisalaam S, Su F, Raj P, Dozmorov I, et al. , (2017). RAD51 interconnects between DNA replication, DNA repair and immunity. Nucleic Acids Res. 45, 4590–4605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Zhang X, Brann TW, Zhou M, Yang J, Oguariri RM, Lidie KB, et al. , (2011). Cutting edge: Ku70 is a novel cytosolic DNA sensor that induces type III rather than type I IFN. J. Immunol. 186, 4541–4545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Ferguson BJ, Mansur DS, Peters NE, Ren H, Smith GL, (2012). DNA-PK is a DNA sensor for IRF-3dependent innate immunity. Elife 1, e00047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Sui H, Zhou M, Imamichi H, Jiao X, Sherman BT, Lane HC, et al. , (2017). STING is an essential mediator of the Ku70-mediated production of IFN-lambda1 in response to exogenous DNA. Sci Signal. 10 [DOI] [PubMed] [Google Scholar]
- 152.Sui H, Chen Q, Imamichi T, (2021). Cytoplasmic-translocated Ku70 senses intracellular DNA and mediates interferon-lambda1 induction. Immunology 163, 323–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Burleigh K, Maltbaek JH, Cambier S, Green R, Gale M Jr., James RC, et al. , (2020). Human DNA-PK activates a STING-independent DNA sensing pathway. Sci. Immunol. 5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Karpova AY, Trost M, Murray JM, Cantley LC, Howley PM, (2002). Interferon regulatory factor-3 is an in vivo target of DNA-PK. Proc. Natl. Acad. Sci. U. S. A. 99, 2818–2823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wang Y, Fu Z, Li X, Liang Y, Pei S, Hao S, et al. , (2021). Cytoplasmic DNA sensing by KU complex in aged CD4(+) T cell potentiates T cell activation and aging-related autoimmune inflammation. Immunity 54 632–47 e9. [DOI] [PubMed] [Google Scholar]
- 156.Gould SJ, Lewontin RC, (1979). The spandrels of San Marco and the Panglossian paradigm: a critique of the adaptationist programme. Proc. R. Soc. Lond. B Biol. Sci. 205, 581–598. [DOI] [PubMed] [Google Scholar]
- 157.Garcia G Jr., Sharma A, Ramaiah A, Sen C, Purkayastha A, Kohn DB, et al. , (2021). Antiviral drug screen identifies DNA-damage response inhibitor as potent blocker of SARS-CoV-2 replication. Cell Rep. 35, 108940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Fakhry C, Westra WH, Li S, Cmelak A, Ridge JA, Pinto H, et al. , (2008). Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J. Natl Cancer Inst. 100, 261–269. [DOI] [PubMed] [Google Scholar]
- 159.Shaheen M, Allen C, Nickoloff JA, Hromas R, (2011). Synthetic lethality: exploiting the addiction of cancer to DNA repair. Blood 117, 6074–6082. [DOI] [PubMed] [Google Scholar]
- 160.Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al. , (2005). Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434, 913–917. [DOI] [PubMed] [Google Scholar]
- 161.Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, et al. , (2009). Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 361, 123–134. [DOI] [PubMed] [Google Scholar]
- 162.Pu X, Wang Z, Klaunig JE, (2015). Alkaline Comet Assay for Assessing DNA Damage in Individual Cells. Curr. Protoc. Toxicol. 65 3.12.1–3.12.11. [DOI] [PubMed] [Google Scholar]
- 163.Kawashima Y, Yamaguchi N, Teshima R, Narahara H, Yamaoka Y, Anai H, et al. , (2017). Detection of DNA double-strand breaks by pulsed-field gel electrophoresis. Genes Cells 22, 84–93. [DOI] [PubMed] [Google Scholar]
- 164.Gunn A, Bennardo N, Cheng A, Stark JM, (2011). Correct end use during end joining of multiple chromosomal double strand breaks is influenced by repair protein RAD50, DNA-dependent protein kinase DNA-PKcs, and transcription context. J. Biol. Chem. 286, 42470–42482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Gunn A, Stark JM, (2012). I-SceI-based assays to examine distinct repair outcomes of mammalian chromosomal double strand breaks. Methods Mol. Biol. 920, 379–391. [DOI] [PubMed] [Google Scholar]
- 166.Richardson C, Moynahan ME, Jasin M, (1998). Double-strand break repair by interchromosomal recombination: suppression of chromosomal translocations. Genes Dev. 12, 3831–3842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Schumacher AJ, Mohni KN, Kan Y, Hendrickson EA, Stark JM, Weller SK, (2012). The HSV-1 exonuclease, UL12, stimulates recombination by a single strand annealing mechanism. PLoS Pathog. 8, e1002862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Richardson CD, Kazane KR, Feng SJ, Zelin E, Bray NL, Schafer AJ, et al. , (2018). CRISPR-Cas9 genome editing in human cells occurs via the Fanconi anemia pathway. Nature Genet. 50, 1132–1139. [DOI] [PubMed] [Google Scholar]
- 169.Roidos P, Sungalee S, Benfatto S, Sercin O, Stutz AM, Abdollahi A, et al. , (2020). A scalable CRISPR/Cas9-based fluorescent reporter assay to study DNA double-strand break repair choice. Nature Commun. 11, 4077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Glaser A, McColl B, Vadolas J, (2016). GFP to BFP Conversion: A Versatile Assay for the Quantification of CRISPR/Cas9-mediated Genome Editing. Mol. Ther. Nucleic Acids 5, e334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Pujals A, Favre L, Pioche-Durieu C, Robert A, Meurice G, Le Gentil M, et al. , (2015). Constitutive autophagy contributes to resistance to TP53-mediated apoptosis in Epstein-Barr virus-positive latency III B-cell lymphoproliferations. Autophagy 11, 2275–2287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Wang L, Howell MEA, McPeak B, Riggs K, Kohne C, Yohanon JU, et al. , (2018). LIMD1 is induced by and required for LMP1 signaling, and protects EBV-transformed cells from DNA damage-induced cell death. Oncotarget 9, 6282–6297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Gruhne B, Sompallae R, Masucci MG, (2009). Three Epstein-Barr virus latency proteins independently promote genomic instability by inducing DNA damage, inhibiting DNA repair and inactivating cell cycle checkpoints. Oncogene 28, 3997–4008. [DOI] [PubMed] [Google Scholar]
- 174.Piekna-Przybylska D, Sharma G, Maggirwar SB, Bambara RA, (2017). Deficiency in DNA damage response, a new characteristic of cells infected with latent HIV-1. Cell Cycle 16, 968–978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Piekna-Przybylska D, Maggirwar SB, (2018). CD4+ memory T cells infected with latent HIV-1 are susceptible to drugs targeting telomeres. Cell Cycle 17, 2187–2203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Stanley SK, Folks TM, Fauci AS, (1989). Induction of expression of human immunodeficiency virus in a chronically infected promonocytic cell line by ultraviolet irradiation. AIDS Res. Hum. Retroviruses 5, 375–384. [DOI] [PubMed] [Google Scholar]
- 177.Li C, Mousseau G, Valente ST, (2019). Tat inhibition by didehydro-Cortistatin A promotes heterochromatin formation at the HIV-1 long terminal repeat. Epigenetics Chromatin. 12, 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Kumar A, Darcis G, Van Lint C, Herbein G, (2015). Epigenetic control of HIV-1 post integration latency: implications for therapy. Clin. Epigenetics 7, 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Duggal NK, Emerman M, (2012). Evolutionary conflicts between viruses and restriction factors shape immunity. Nature Rev. Immunol. 12, 687–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Demogines A, East AM, Lee JH, Grossman SR, Sabeti PC, Paull TT, et al. , (2010). Ancient and recent adaptive evolution of primate non-homologous end joining genes. PLoS Genet. 6, e1001169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Lou DI, McBee RM, Le UQ, Stone AC, Wilkerson GK, Demogines AM, et al. , (2014). Rapid evolution of BRCA1 and BRCA2 in humans and other primates. BMC Evol. Biol. 14, 155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Petrucelli N, Daly MB, Feldman GL, (2010). Hereditary breast and ovarian cancer due to mutations in BRCA1 and BRCA2. Genet. Med. 12, 245–259. [DOI] [PubMed] [Google Scholar]
- 183.Xu LH, Huang M, Fang SG, Liu DX, (2011). Coronavirus infection induces DNA replication stress partly through interaction of its nonstructural protein 13 with the p125 subunit of DNA polymerase delta. J. Biol. Chem. 286, 39546–39559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Li R, Wang L, Liao G, Guzzo CM, Matunis MJ, Zhu H, et al. , (2012). SUMO binding by the Epstein-Barr virus protein kinase BGLF4 is crucial for BGLF4 function. J. Virol. 86, 5412–5421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Choudhuri T, Verma SC, Lan K, Murakami M, Robertson ES, (2007). The ATM/ATR signaling effector Chk2 is targeted by Epstein-Barr virus nuclear antigen 3C to release the G2/M cell cycle block. J. Virol. 81, 6718–6730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Sato Y, Kamura T, Shirata N, Murata T, Kudoh A, Iwahori S, et al. , (2009). Degradation of phosphorylated p53 by viral protein-ECS E3 ligase complex. PLoS Pathog. 5, e1000530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Kudoh A, Fujita M, Zhang L, Shirata N, Daikoku T, Sugaya Y, et al. , (2005). Epstein-Barr virus lytic replication elicits ATM checkpoint signal transduction while providing an S-phase-like cellular environment. J. Biol. Chem. 280, 8156–8163. [DOI] [PubMed] [Google Scholar]
- 188.Chen YR, Liu MT, Chang YT, Wu CC, Hu CY, Chen JY, (2008). Epstein-Barr virus latent membrane protein 1 represses DNA repair through the PI3K/Akt/ FOXO3a pathway in human epithelial cells. J. Virol. 82, 8124–8137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Knight JS, Sharma N, Robertson ES, (2005). SCFSkp2 complex targeted by Epstein-Barr virus essential nuclear antigen. Mol. Cell. Biol. 25, 1749–1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
