Fact-checked by Grok 2 weeks ago

DNA-PKcs

DNA-dependent catalytic subunit () is a large / belonging to the phosphatidylinositol 3-kinase-related kinase (PIKK) family, encoded by the PRKDC gene (also known as XRCC7) on human 8q11. With a molecular mass of approximately 469 kDa and consisting of 4,129 , it forms the catalytic of the DNA-PK holoenzyme, which plays a central role in repairing DNA double-strand breaks (DSBs) via the (NHEJ) pathway. This pathway is critical for maintaining genomic stability, as DSBs represent one of the most severe forms of DNA damage induced by , chemotherapeutic agents, or replication stress. The DNA-PK activity was first observed in 1985, when double-stranded DNA was shown to stimulate the phosphorylation of several HeLa cell nuclear proteins, including poly(ADP-ribose) polymerase, revealing a novel DNA-activated kinase system. In 1990, the catalytic subunit was purified and identified from HeLa cell nuclei, confirming DNA-PKcs as a DNA-end-activated protein kinase distinct from other known kinases. Structurally, DNA-PKcs features an N-terminal region rich in HEAT repeats that form a flexible circular cradle for protein interactions, a central head unit containing the kinase domain, and C-terminal FAT (FRAP-ATM-TRRAP) and FATC domains that regulate kinase activity. Upon DNA damage, the Ku70/Ku80 heterodimer binds to DSB ends, recruits DNA-PKcs to form the holoenzyme, and activates its autophosphorylation and substrate phosphorylation capabilities, targeting over 700 proteins involved in repair. Beyond DSB repair, DNA-PKcs contributes to V(D)J recombination during T- and B-cell development, telomere end protection, and cell cycle checkpoints at G1/S and G2/M phases. It also participates in responses to replication stress, autophagy induction, and cellular senescence, with nuclear and cytoplasmic localization enabling diverse regulatory roles. Mutations in PRKDC lead to severe combined immunodeficiency in animal models and human inflammatory diseases characterized by granulomas, autoimmunity, and recurrent infections. In cancer, DNA-PKcs overexpression promotes tumor survival and resistance to radiotherapy and chemotherapy, positioning it as a promising therapeutic target for inhibitors like NU7441 and M3814.

Discovery and Structure

Historical Discovery

The discovery of DNA-dependent (DNA-PK) activity occurred serendipitously in 1985 during experiments on in cell extracts. Researchers observed that double-stranded DNA stimulated the of several proteins, including a prominent 350-400 kDa polypeptide, revealing a novel DNA-activated serine/ involved in the cellular response to DNA damage. In 1990, independent studies identified the catalytic subunit of this , later termed DNA-PKcs, through biochemical purification from human cell extracts. Carter et al. isolated a ~350 kDa protein that exhibited DNA-dependent autophosphorylation and phosphorylated substrates such as the Hsp90, confirming its role as the enzymatic core. Concurrently, Lees-Miller et al. demonstrated that this subunit phosphorylated the tumor suppressor at specific serine residues, linking it to DNA damage signaling pathways via assays with purified fractions. These findings established DNA-PKcs as a distinct entity separate from the DNA-binding component later identified as Ku. The cloning of the PRKDC gene encoding DNA-PKcs was achieved in 1995 by Hartley et al., who sequenced the full-length cDNA from human cells, revealing a massive 4,128-amino-acid protein of approximately 469 kDa with homology to phosphatidylinositol 3-kinase family members. This molecular characterization enabled targeted studies, including the pivotal demonstration that mutations in the murine Prkdc gene cause severe combined immunodeficiency (SCID) in mice. Kirchgessner et al. showed that SCID fibroblasts and lymphocytes lacked DNA-PK activity due to defects in the catalytic subunit, directly tying DNA-PKcs to V(D)J recombination and double-strand break repair. Contributions from the laboratories of Graeme C.M. Smith and Stephen P. Jackson were instrumental in defining the DNA-PK holoenzyme composition, integrating DNA-PKcs with the Ku70/Ku80 heterodimer through immunoprecipitation and reconstitution assays.

Molecular Structure

DNA-PKcs, the catalytic subunit of the DNA-dependent complex, is encoded by the PRKDC gene located on human 8q11 and consists of 4,128 , yielding a of approximately 469 kDa. The domain architecture of DNA-PKcs features an extensive N-terminal region of repeats (residues 1–2801) that folds into an open, ring-like scaffold facilitating regulatory interactions and conformational flexibility. This is followed by the domain (residues 2802–3564), which stabilizes the adjacent catalytic regions. The domain (residues 3565–4100) encompasses conserved motifs, including the ATP-binding site and catalytic loop, essential for substrate . The short C-terminal FATC domain (residues 4101–4128) maintains autoinhibition in the apo state by interacting with the kinase domain, a constraint relieved upon complex activation.30907-2) High-resolution cryo-EM structures from 2022 and 2023 have elucidated the conformational transitions of DNA-PKcs, revealing a compact, closed (autoinhibited) form in isolation and an extended, open (activated) state upon engagement with DNA-bound . These structures demonstrate how DNA-induced stretching and twisting of the repeats propagate to the head domain, repositioning the for catalysis.30907-2) Within the DNA-PK holoenzyme, DNA-PKcs integrates with the Ku70/Ku80 heterodimer, where double-strand DNA ends serve as a bridge to stabilize the assembly and align the components for .30907-2)

Mechanism of Action

Activation and Kinase Activity

The of DNA-PKcs, the catalytic subunit of the DNA-dependent protein kinase complex, is tightly regulated and occurs primarily at DNA double-strand break (DSB) ends in a process dependent on the Ku70/Ku80 heterodimer. The Ku heterodimer initially binds to the DSB ends with high affinity, forming a ring structure that slides along the DNA and recruits DNA-PKcs through interactions with its N-terminal repeats and circular cradle . This recruitment positions DNA-PKcs such that the DNA end docks beneath its head , inducing a conformational change that transitions the enzyme from an autoinhibited, closed state to an open, active configuration, thereby releasing the autoinhibitory interaction between the FAT and the kinase . This synapsis-dependent is further stabilized by ATP binding, which promotes dissociation of inhibitory elements and enables subsequent autophosphorylation. Central to this activation is the autophosphorylation of DNA-PKcs at specific sites within clustered motifs, which modulates its accessibility to DNA ends and facilitates downstream repair processes. Key autophosphorylation occurs in the ABCDE cluster (residues 2609–2647), including sites such as Thr2609, Ser2612, Thr2620, Ser2624, Thr2638, and Thr2647; phosphorylation here, particularly at Thr2609 and Thr2620, induces structural rearrangements that open the DNA-binding cleft, allowing end processing. These events are cis-autophosphorylation within the but can involve trans-phosphorylation in dimeric complexes for other clusters like PQR (residues 2023–2056). Recent cryo-EM structures from 2022 reveal that in the active state, the DNA-end-binding (DEB) helix (residues 2735–2768) interacts directly with the DNA terminus and the ABCDE cluster, constraining DNA movement and fine-tuning activation to prevent premature dissociation. As a serine/threonine , DNA-PKcs catalyzes the transfer of the γ-phosphate from ATP to target , exhibiting a preference for the consensus motif S/T-Q (serine or followed by ), often extended as S/T-Q-X-E or involving prior phospho-serine/. This motif is evident in its autophosphorylation sites and like XRCC4 and , enabling coordinated events during repair. activity is typically assayed using as a model , where is quantified by incorporation of radiolabeled ATP or luminescent detection of ADP production. Kinetic studies indicate a Km for ATP of approximately 18 μM under optimized conditions with Mg²⁺, reflecting its efficiency at physiological levels.

Role in DNA Repair Pathways

DNA-PKcs functions as a core component of the non-homologous end joining (NHEJ) pathway, the primary mechanism for repairing DNA double-strand breaks (DSBs) in mammalian cells, particularly during the G1 phase of the cell cycle. Upon DSB induction, the Ku70/Ku80 heterodimer initially binds to the free DNA ends, serving as a scaffold that recruits DNA-PKcs to form the active DNA-PK holoenzyme. This assembly promotes DNA end synapsis by tethering the broken ends in close proximity, which facilitates the subsequent recruitment of the XRCC4-DNA ligase IV (LIG4) complex responsible for end ligation, often with limited nucleotide loss or addition to minimize genomic alterations. This process ensures rapid, albeit error-prone, repair to maintain genome stability in non-replicating cells. In DSB sensing and initial response, DNA-PKcs enhances the stability of the Ku complex at break sites, thereby shielding DNA ends from nucleases and preventing excessive end resection that would otherwise favor (). This protective role inhibits the action of resection-initiating factors such as the MRN complex (MRE11-RAD50-NBS1) and CtIP, preserving blunt or minimally processed ends suitable for direct NHEJ ligation. By doing so, DNA-PKcs enforces pathway fidelity, prioritizing NHEJ over in contexts where are absent. Recent 2024 phosphoproteomic analyses have further uncovered non-canonical signaling by DNA-PKcs, including novel S/T-ψ-D/E motifs. Beyond its canonical NHEJ function, DNA-PKcs exerts regulatory influence on alternative repair pathways, acting as a suppressor of by blocking end resection and thereby serving as a backup to channel unrepaired DSBs away from error-free HR templating. In situations of NHEJ compromise or high cellular stress, such as exposure, DNA-PKcs can contribute to (MMEJ) in certain contexts, while primarily suppressing excessive MMEJ to curb illegitimate chromosomal rearrangements. This pathway hierarchy underscores NHEJ's dominance in G1, where DNA-PKcs activity predominates, contrasted by competitive dynamics with in S/G2 phases, where cyclin-dependent kinase-mediated modifications can tip the balance toward resection and .

Biological Functions

Non-Homologous End Joining

(NHEJ) is the primary pathway for repairing DNA double-strand breaks (DSBs) in mammalian cells, particularly during the G0/G1 phases of the , where it operates in a template-independent manner to rejoin broken DNA ends with minimal . DNA-PKcs, the catalytic subunit of the DNA-dependent protein kinase (DNA-PK) complex, plays a central and multifaceted role in this classical NHEJ (cNHEJ) process by facilitating end recognition, protection, processing, and ligation, thereby maintaining genomic stability. Defects in DNA-PKcs impair NHEJ efficiency, leading to persistent DSBs and chromosomal instability. The NHEJ pathway initiates with the rapid binding of the Ku70/Ku80 heterodimer () to the free DNA ends at a DSB, forming a ring-like structure that slides inward along the DNA to protect the termini from degradation and nucleate further repair complex assembly. then recruits DNA-PKcs through direct protein-protein interactions, assembling the DNA-PK holoenzyme, which is essential for stabilizing the break and promoting the alignment of DNA ends in a process known as . This tethering function of DNA-PKcs prevents diffusion of the broken ends and sets the stage for subsequent repair steps, with the holoenzyme's activity becoming activated upon DNA binding. Once formed, the DNA-PK holoenzyme undergoes at multiple sites on DNA-PKcs, which induces conformational changes that enable end processing for non-ligatable termini. This activates the endonuclease , which associates with DNA-PKcs to resolve complex structures such as hairpins, 3' overhangs, or damaged through limited nucleolytic resection, typically removing fewer than 20 to generate compatible ends without extensive loss of genetic information. The processed ends are then bridged by DNA-PKcs-mediated tethering to align them for . The final stage involves of the ligation machinery, including the XRCC4/LIG4 complex and the accessory factor XLF (also known as XRCC4-like factor), which form a multi-subunit scaffold to seal the nick between the processed DNA ends. DNA-PKcs facilitates this and, upon successful , undergoes further and dissociation from the DNA, allowing completion of repair and release of the core components. In mammalian cells, NHEJ mediated by DNA-PKcs repairs approximately 80% of DSBs, underscoring its dominance over alternative pathways, though inefficiencies or defects in this process result in chromosomal aberrations and heightened . Recent investigations as of 2024 have elucidated an additional protective role for DNA-PKcs in NHEJ, where the holoenzyme shields "clean" blunt or minimally overhang-ended DSBs from resection by exonucleases such as Exo1 or Mre11, thereby favoring rapid and faithful rejoining over error-prone or alternative end-joining pathways. This end-blocking function, mediated by structural elements like the DNA end-blocking helix in DNA-PKcs, enhances repair fidelity for undamaged breaks, with implications for therapeutic targeting in .

V(D)J Recombination

V(D)J recombination is a site-specific process in developing B and T lymphocytes that assembles variable (V), diversity (D), and joining (J) gene segments to generate diverse receptors essential for adaptive immunity. The recombination-activating genes and RAG2 form a complex that recognizes recombination signal sequences () flanking the V, D, and J segments, adhering to the 12/23 rule where a 12-base pair spacer RSS pairs only with a 23-base pair spacer RSS. RAG1/2 introduces double-strand breaks (DSBs) at the RSS-coding segment borders through nicking and transesterification, yielding blunt signal ends (SEs) attached to RSS and covalently sealed hairpin coding ends (CEs). DNA-PKcs, as the catalytic subunit of the (DNA-PK) holoenzyme, is recruited to these DSBs by the Ku70/Ku80 heterodimer, which initially binds the DNA ends to form a scaffold for (NHEJ). In the joining phase, DNA-PKcs facilitates the formation of signal joints (SJs) and coding joints (CJs). Signal ends, being blunt, are ligated directly by the NHEJ ligase complex (XRCC4-LIG4-XLF) with minimal processing, often forming non-replicative extrachromosomal circles. Coding ends require more extensive processing due to their hairpin structure; DNA-PKcs phosphorylates and activates the endonuclease, enabling it to open the hairpins asymmetrically and generate 3' overhangs that undergo nucleotide additions by (TdT) or exonucleolytic nibbling. This processing, coordinated by DNA-PKcs, ensures precise joining of coding segments while introducing variability at junctions. Genetic studies underscore the essential role of DNA-PKcs in V(D)J recombination. Mutations in the Prkdc gene encoding DNA-PKcs, as observed in severe combined immunodeficient (scid) mice, abolish V(D)J recombination by preventing hairpin opening and joint formation, resulting in a complete block of B and T cell development. Similar defects occur in human patients with DNA-PKcs deficiencies, such as radiosensitive T-B- SCID, confirming its indispensability. The fidelity of V(D)J recombination, supported by DNA-PKcs, generates junctional diversity through random nucleotide deletions and additions at CE junctions, contributing to an estimated 10^{11} to 10^{18} unique antigen receptor specificities theoretically in humans, with the realized repertoire around 10^9. This diversity is crucial for recognizing a vast array of pathogens while maintaining recombination accuracy to avoid genomic instability. DNA-PKcs also plays an essential role in class switch recombination (CSR) in mature B cells, where activation-induced cytidine deaminase (AID) generates DSBs in switch regions, and DNA-PKcs facilitates their repair via NHEJ to enable antibody isotype switching. Defects in DNA-PKcs impair CSR, leading to immunodeficiencies.

Regulation

Protein Interactions

DNA-PKcs forms the core of the DNA-dependent protein kinase (DNA-PK) complex through direct binding to the Ku70/Ku80 heterodimer, which recognizes and binds DNA double-strand breaks to recruit DNA-PKcs via interactions with its N-terminal HEAT repeats. This stable association positions DNA-PKcs at DNA ends, enabling its kinase activation and initiation of non-homologous end joining (NHEJ) repair. Following autophosphorylation, DNA-PKcs facilitates the recruitment of XRCC4 and DNA ligase IV (LIG4), which bind at DNA termini to promote ligation; this post-phosphorylation step enhances ligase activity and complex stability. Phosphorylation effects on these interactions further modulate downstream assembly. Beyond the core NHEJ machinery, DNA-PKcs engages regulatory partners that influence its stability and signaling. Heat shock protein 90 () interacts with DNA-PKcs to chaperone its folding and maintain protein stability, as HSP90 inhibition reduces DNA-PKcs levels and impairs . Similarly, DNA-PKcs binds , phosphorylating it to regulate and responses, establishing crosstalk between and tumor suppression pathways. Recent mass spectrometry-based studies have mapped non-canonical interactors, revealing associations with metabolic enzymes such as those in , which modulate enzymatic activity and energy production under DNA damage conditions. These interactions exhibit distinct dynamics: the Ku70/Ku80 binding is relatively stable, stabilizing at DNA ends for several minutes to allow repair initiation, whereas associations like those with are more transient, occurring post-activation to facilitate end processing and dissociation. Such temporal regulation prevents prolonged occupancy that could block repair progression. Yeast two-hybrid screening and co-immunoprecipitation (co-IP) assays have been instrumental in identifying over 50 protein interactors of DNA-PKcs, mapping binding domains and validating functional complexes and in cells.

Post-Translational Modifications

DNA-PKcs undergoes extensive post-translational modifications, predominantly , which dynamically regulate its conformation, activity, and interactions during DNA damage response. These modifications are critical for controlling access to DNA ends and facilitating the progression of (NHEJ). occurs at multiple sites across the protein, with over 40 identified, including key clusters that respond to double-strand breaks induced by or other genotoxic stresses. Autophosphorylation is a primary regulatory mechanism for DNA-PKcs, enabling conformational changes that promote DNA end and complex disassembly. The ABCDE cluster, encompassing residues Thr2609 to Thr2647 (including sites Thr2609, Ser2612, Thr2620, Ser2624, Thr2638, and Thr2647), is autophosphorylated upon DNA-PK , facilitating the opening of the DNA-PK holoenzyme to allow for downstream repair factors like and ligase IV. In contrast, autophosphorylation at the PQR cluster (Ser2023 to Ser2053, including Ser2056) promotes the disassembly of the DNA-PK complex post-ligation, preventing prolonged retention at repair sites and ensuring repair . These clustered phosphorylations are interdependent, with ABCDE modifications often preceding PQR events to coordinate the repair cycle. Hetero-phosphorylation by related kinases further fine-tunes DNA-PKcs function, particularly in checkpoint activation. ATM phosphorylates DNA-PKcs at Ser2056 and the Thr2609 cluster, while ATR phosphorylates DNA-PKcs under conditions of replication stress, enhancing intra-S-phase checkpoints and coordinating with pathways. For instance, -mediated phosphorylation at Thr2609 stabilizes DNA-PKcs at damage sites, amplifying signaling for arrest. Recent phosphoproteomic analyses have expanded the understanding of DNA-PKcs modifications, identifying dozens of additional sites and linking them to non-canonical signaling beyond classical NHEJ. A 2024 study mapped over 1,200 conserved events in response to , revealing DNA-PKcs-dependent motifs (e.g., S/T-ψ-D/E) that extend its substrate specificity to transcription and replication factors, thereby influencing broader cellular responses like . Functional impacts of these modifications are evident in studies; for example, non-phosphorylatable mutants at the ABCDE cluster (e.g., 6A variant) impair end processing and increase , while promotes efficient repair but requires precise for resolution.

Physiological Roles

Immunity and Development

DNA-PKcs plays a critical role in lymphocyte development by facilitating V(D)J recombination, which is essential for the maturation of B and T cells in the adaptive immune system. Defects in DNA-PKcs lead to impaired V(D)J recombination, resulting in severe combined immunodeficiency (SCID) phenotypes characterized by a profound block in B and T cell development, as observed in DNA-PKcs-deficient mouse models. In these models, the absence of functional DNA-PKcs causes accumulation of unrepaired DNA double-strand breaks during recombination, preventing the generation of diverse antigen receptors and halting lymphocyte differentiation. Beyond V(D)J recombination, DNA-PKcs supports class switch recombination (CSR) in mature B cells, enabling the switching of immunoglobulin heavy chain constant regions from IgM to other isotypes like IgG, IgA, or IgE through non-homologous end joining (NHEJ) repair of activation-induced cytidine deaminase-generated double-strand breaks. Loss of DNA-PKcs significantly reduces CSR efficiency to most isotypes, except IgG1 in some contexts, leading to altered antibody responses and impaired humoral immunity. This NHEJ-dependent function underscores DNA-PKcs's necessity for B cell plasticity in responding to diverse pathogens. In non-immune developmental processes, DNA-PKcs contributes to neural development by promoting the survival of newly generated neurons in the embryonic brain. DNA-PKcs-deficient mice exhibit elevated DNA double-strand breaks and increased in the , particularly in developing neurons, leading to subtle cerebellar defects and impaired without causing embryonic lethality. These findings highlight DNA-PKcs's broader role in maintaining genomic stability during tissue development outside the . Recent studies as of 2025 have revealed DNA-PKcs's involvement in innate immunity through modulation of cytokine signaling pathways, including the cGAS-STING axis, where it influences interferon and inflammatory cytokine production in response to cytosolic DNA. This function links DNA-PKcs to antiviral innate responses and broader inflammatory regulation during development and homeostasis.

Telomere Maintenance

DNA-PKcs plays a crucial role in telomere protection by being recruited to ends through interactions with the complex components TRF2 and RAP1. This recruitment forms a TRF2/RAP1-DNA-PK complex that represses classical (c-NHEJ) at s, thereby preventing deleterious end-to-end fusions that could lead to genomic instability. The complex specifically inhibits the end-joining function of DNA-PK by blocking the recruitment of XRCC4-LIG4 to Ku-bound DNA ends, ensuring that telomeric DNA remains protected from inappropriate repair. This mechanism complements other -mediated protections, such as t-loop formation, to maintain integrity during the . A key 2025 discovery revealed the structural basis of this repression, showing that RAP1 directly interacts with the Ku heterodimer within the DNA-PK holoenzyme, occluding the LIG4 and thereby inhibiting end at telomeres. Cryo-electron microscopy structures at 3.58 Å resolution confirmed that the Myb and BRCT domains of RAP1 engage specific domains on Ku70, stabilizing the complex while preventing downstream NHEJ progression. This RAP1-mediated inhibition maintains telomere capping, particularly at blunt leading-strand ends or shortened telomeres where repair pathways might otherwise be activated. Without this regulation, telomeres become vulnerable to fusion events, as evidenced by increased chromosomal end-to-end joins in cells lacking RAP1 or TRF2. In addition to repressing c-NHEJ, DNA-PKcs suppresses (MMEJ, also known as alternative NHEJ) at short or deprotected , limiting error-prone repairs that exacerbate telomere attrition. This suppression helps avert the activation of persistent DNA damage responses that drive replicative , as MMEJ-dependent fusions at critically short ends can propagate genomic rearrangements. Studies indicate that DNA-PKcs promotes end stability to favor protective capping over mutagenic MMEJ, particularly in contexts of telomere shortening during prolonged . Evidence from DNA-PKcs-deficient models underscores its essential function in telomere maintenance. In embryonic fibroblasts, loss of DNA-PKcs results in telomere uncapping, manifested as elevated rates of end-to-end fusions and chromosomal fragments, leading to through breakage-fusion-bridge cycles. Conditional ablation approaches further demonstrate that tissue-specific DNA-PKcs deficiency induces telomere dysfunction and associated chromosomal instability, highlighting its non-redundant role in preventing in proliferative cells. These findings establish DNA-PKcs as a key guardian of ends against repair-mediated damage.

Pathological Implications

Cancer

Dysregulation of DNA-PKcs plays a significant role in cancer development and progression, primarily through its involvement in DNA double-strand break repair via (NHEJ), which allows tumor cells to survive genotoxic stress. Overexpression of DNA-PKcs is frequently observed in various malignancies, including , , non-small cell , gastric, ovarian, pancreatic, and hepatocellular cancers, where it enhances cell survival and resistance to DNA-damaging therapies by promoting efficient NHEJ-mediated repair. Genetic alterations in the PRKDC gene encoding DNA-PKcs occur in more than 10% of numerous cancer types and can contribute to genomic instability by impairing fidelity, thereby facilitating tumor evolution and heterogeneity. Beyond its canonical repair functions, DNA-PKcs exerts non-canonical effects that drive tumorigenesis, as highlighted in a 2024 review. Its kinase activity, independent of , promotes metabolic reprogramming by phosphorylating and activating glycolytic enzymes such as (ALDOA) and pyruvate kinase M2 (PKM2), thereby enhancing and supporting tumor growth in castration-resistant . Additionally, DNA-PKcs facilitates cancer cell migration and through phosphorylation of transcription factors like Snail1, which increases and genomic instability, and by upregulating (VEGF) expression to promote and . These kinase-dependent mechanisms contribute to protumorigenic signaling in multiple cancer contexts. High DNA-PKcs expression in tumors sensitizes them to radiosensitization strategies, as inhibiting its activity impairs NHEJ and exacerbates DNA damage from , particularly in . In tumors with BRCA1/2 deficiencies, where is compromised, DNA-PKcs becomes critical for NHEJ-dependent survival, making it a key mediator of resistance to therapies like ; its upregulation in such contexts further promotes tumor persistence. Clinically, elevated DNA-PKcs levels correlate with poor prognosis in patients, associating with reduced survival following standard treatments including radiation.

Immunodeficiency and Disease

Mutations in the PRKDC gene, which encodes DNA-PKcs, cause rare forms of severe combined immunodeficiency (SCID), characterized by profound defects in T- and B-cell development due to impaired V(D)J recombination and non-homologous end joining (NHEJ). These mutations lead to radiosensitive SCID (RS-SCID), with affected individuals exhibiting hypersensitivity to ionizing radiation, recurrent infections, and failure to thrive. For instance, the recurrent p.L3062R missense mutation has been identified in multiple patients, resulting in atypical SCID phenotypes including low but detectable T-cell counts and neurological abnormalities such as microcephaly, seizures, and developmental delay. Growth retardation is a common feature, often accompanied by dysmorphic features and increased susceptibility to granulomatous disease. The prevalence of SCID overall is estimated at 1 in 50,000 to 100,000 live births, but PRKDC-related cases are exceptionally rare, with fewer than 20 human patients reported to date. models, including the classical scid with a truncating Prkdc and complete DNA-PKcs (DNA-PKcs^{-/-}) lines, closely recapitulate these human traits, displaying absent adaptive immunity, , and growth defects, though without the severe neurological involvement seen in some human cases. These models have been instrumental in elucidating the role of DNA-PKcs in immune development and . Beyond SCID, DNA-PKcs dysfunction has been linked to other immunodeficiencies and autoimmune disorders, including associations with through defects in immunoglobulin class-switch recombination (CSR). Impaired CSR in DNA-PKcs-deficient B cells leads to reduced production of switched isotypes (IgG, IgA, IgE), contributing to humoral immune defects and potentially exacerbating autoimmune inflammation in conditions like . Recent 2025 studies in DNA-PKcs knockout mice have further revealed neurodegeneration phenotypes, including impaired synaptic transmission, deficits in , and accumulation of DNA damage in neurons, suggesting a broader role in maintenance. In the context of aging, accumulated double-strand breaks (DSBs) over time contribute to DNA-PKcs exhaustion, where chronic activation of the leads to diminished repair capacity and . DNA-PKcs plays a protective role at , and its deficiency promotes telomere uncapping, triggering persistent signaling and (SASP), which accelerates tissue aging and age-related immune decline. This exhaustion mechanism underscores DNA-PKcs as a key mediator linking genomic instability to .

Therapeutic Targeting

DNA-PKcs Inhibitors

Small-molecule inhibitors of DNA-PKcs primarily target its kinase domain to disrupt non-homologous end joining (NHEJ) repair of DNA double-strand breaks (DSBs). These inhibitors are classified mainly as ATP-competitive agents that bind the ATP-binding pocket, preventing phosphorylation events essential for DNA repair. Early examples include NU7441, a chromen-4-one derivative with a biochemical IC50 of 14 nM against DNA-PKcs, which demonstrates modest selectivity over related PI3K family kinases (IC50 of 5 μM for PI3K). More advanced inhibitors, such as M3814 (peposertib), also operate via ATP competition, achieving a biochemical IC50 of 0.6 nM at low ATP concentrations and showing greater than 100-fold selectivity over PI3K isoforms (e.g., IC50 of 95 nM for PI3Kδ). Although some structural analyses suggest potential allosteric modulation in related inhibitors, M3814 and NU7441 are confirmed as ATP-competitive based on ATP-dependent potency shifts. The primary mechanism of these inhibitors involves blocking the activity of DNA-PKcs, which halts autophosphorylation at key sites like Ser2056 and impairs downstream end-processing and in NHEJ. This leads to accumulation of unrepaired DSBs, inducing genomic instability and in response to DNA-damaging agents. , DNA-PKcs inhibitors enhance (IR) sensitivity by 2- to 5-fold, as measured by dose enhancement factors in clonogenic survival assays across lines such as A549 and , without significantly affecting normal cell repair at low doses. For instance, NU7441 prevents IR-induced autophosphorylation and increases persistent γH2AX foci, potentiating DSB persistence. AZD7648, a 7,9-dihydro-8H-purin-8-one with a biochemical IC50 of 0.6 nM and over 1,000-fold selectivity against PI3Kγ (IC50 >1 μM), was investigated in phase I/II trials but discontinued in 2025 due to greater-than-expected and limited antitumor activity in advanced tumors. It blocks autophosphorylation and induces genomic instability, showing radiosensitization in preclinical models. Off-target effects remain a consideration; NU7441 exhibits mild PI3K inhibition at higher doses, potentially contributing to , while M3814 and AZD7648 minimize this through optimized scaffolds. Preclinical absorption, distribution, metabolism, and excretion () profiles support oral for clinical translation. In phase I trials, M3814 demonstrates rapid (tmax 1.1–2.5 hours), a of approximately 5.5 hours, and dose-proportional exposure with minimal accumulation upon repeated dosing, enabling once- or twice-daily regimens. Similar favorable are reported for AZD7648 in xenograft models, with good tumor penetration and low systemic toxicity. These properties underpin their potential in enhancing and without exacerbating normal tissue damage in preclinical settings.

Clinical Applications

DNA-PKcs modulation has emerged as a promising strategy in cancer therapy, particularly through that enhance the efficacy of DNA-damaging treatments. Peposertib (M3814), a selective DNA-PKcs , is under in a phase I trial (NCT04555577) combining it with radiotherapy followed by for newly diagnosed MGMT-unmethylated . As of November 2025, data from 21 patients show favorable safety, with reduced DNA-PK phosphorylation in tumors observed. Early data indicate potential radiosensitization without excessive toxicity, though objective response rates remain under evaluation. Additionally, combinations of DNA-PKcs with are being explored in phase I trials for homologous recombination-deficient (HRD) tumors, such as peposertib with pegylated liposomal (NCT04092270), which is active but not recruiting as of September 2025, aiming to exploit by impairing alternative pathways. In gene editing applications, DNA-PKcs inhibition facilitates () over (), improving precision in therapeutic CRISPR-based corrections. The inhibitor AZD7648 has been shown to boost HDR efficiency to approximately 60% at the CD40LG locus in mobilized peripheral blood + hematopoietic and progenitor cells (HSPCs), enabling effective correction of X-linked hyper-IgM , a form of (SCID). This approach reduces NHEJ-mediated insertions/deletions (indels) by up to 13-fold while enhancing long-term engraftment in models. However, despite its preclinical promise, concerns about large-scale genomic alterations like megabase deletions have been noted, and a December 2024 study highlighted dangerous side effects of AZD7648 in gene editing, including increased genomic instability risks across cell types. Beyond and , DNA-PKcs inhibitors show potential in enhancing by promoting immunogenic cell death and antitumor immune responses. For instance, AZD7648 combined with radiotherapy induces in tumor cells, activating the STING pathway and increasing T-cell infiltration, which synergizes with PD-1 checkpoint inhibitors to improve outcomes in preclinical models. Emerging explorations include telomere-targeted strategies leveraging DNA-PKcs's role in end protection, with preliminary links to Hutchinson-Gilford progeria syndrome where disrupts DNA-PKcs function and accelerates attrition, suggesting modulation could mitigate . Clinical translation faces challenges, including potential toxicity such as myelosuppression observed in early trials of DNA-PKcs inhibitors combined with DNA-damaging agents, though single-agent peposertib has shown minimal hematologic adverse events. development, particularly PRKDC expression levels and status, is critical for patient selection, as high PRKDC correlates with resistance to radiotherapy and response in various cancers. Ongoing trials emphasize the need for refined dosing to balance efficacy and safety.

References

  1. [1]
    DNA-PKcs: A Multi-Faceted Player in DNA Damage Response - PMC
    Dec 23, 2020 · DNA-dependent protein kinase catalytic subunit belongs to the PIKKs family, and is a type of DNA-activated serine/threonine protein kinase with ...
  2. [2]
    DNA-Dependent Protein Kinase Catalytic Subunit - NIH
    DNA-PK is thought to act as the “sensor” for DNA double-stranded breaks (DSB), which are considered the most deleterious type of DNA damage.
  3. [3]
    Human DNA-dependent protein kinase catalytic subunit deficiency
    DNA-PKcs deficiency mainly manifests as an inflammatory disease with granuloma and autoimmune features, along with severe infections.
  4. [4]
    DNA-PKcs: A Targetable Protumorigenic Protein Kinase - PMC - NIH
    DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a pleiotropic protein kinase that plays critical roles in cellular processes fundamental to cancer.
  5. [5]
    DNA-dependent protein kinase catalytic subunit: a relative ... - PubMed
    Here, we describe the cloning of the DNA-PKcs cDNA and show that DNA-PKcs falls into the phosphatidylinositol (PI) 3-kinase family. Biochemical ...
  6. [6]
    DNA-dependent kinase (p350) as a candidate gene for the murine ...
    Severe combined immunodeficient (SCID) mice are deficient in a recombination process utilized in both DNA double-strand break repair and in V(D)J recombination.Missing: et al PKcs date
  7. [7]
    PRKDC - DNA-dependent protein kinase catalytic subunit - UniProt
    Involved in DNA non-homologous end joining (NHEJ) required for double-strand break (DSB) repair and V(D)J recombination.
  8. [8]
    PRKDC Gene - Protein Kinase, DNA-Activated, Catalytic Subunit
    This gene encodes the catalytic subunit of the DNA-dependent protein kinase (DNA-PK). It functions with the Ku70/Ku80 heterodimer protein in DNA double strand ...
  9. [9]
    Role of PRKDC in cancer initiation, progression, and treatment - PMC
    Oct 26, 2021 · Structure of PRKDC and DNA-PKcs​​ The DNA-PKcs protein contains 4128 amino acids; it has a relative molecular weight of 469 kDa and is the ...
  10. [10]
    DNA-PKcs: A Multi-Faceted Player in DNA Damage Response
    DNA-activated/or DNA-dependent protein kinase was first reported in 1985 by Walker et al. (1985). They discovered this protein by chance, finding that ...
  11. [11]
    Structural analysis of the basal state of the Artemis:DNA-PKcs complex
    Jul 8, 2022 · Human DNA-PKcs consists of three segments, the N-term (or N-HEAT) region (1–891 aa), the Circular Cradle (or M-HEAT) region (892–2801 aa) and ...Missing: architecture | Show results with:architecture
  12. [12]
    Structure of DNA-PKcs. Panel A - ResearchGate
    The N-HEAT domain is in blue, the M-HEAT/circular cradle is in green, the FAT domain is in purple and the kinase domain is in yellow. Also shown is the ...<|control11|><|separator|>
  13. [13]
    Uncovering DNA-PKcs ancient phylogeny, unique sequence motifs ...
    The PIKK family is composed of six biologically important kinases: 1) the DNA-dependent protein kinase catalytic subunit (DNA-PKcs, gene names PRKDC and XRCC7); ...1.3. Dna-Pkcs... · 1.4. Dna-Pkcs Structure · 3. Results
  14. [14]
    7OTM: Cryo-EM structure of DNA-PKcs in complex with NU7441
    Jan 12, 2022 · Here we report cryo-electron microscopy (cryo-EM) structures of human DNA-PKcs natively purified from HeLa cell nuclear extracts, in complex ...
  15. [15]
    Human DNA-dependent protein kinase activation mechanism - Nature
    Jan 5, 2023 · All PIKKs also have an α-solenoid amino-terminal region, which consists mainly of HEAT repeats that have important regulatory roles. There have ...
  16. [16]
    Cryo-EM structure of the DNA-PK holoenzyme - PNAS
    Jun 26, 2017 · Here, we present cryo-EM structures of the DNA-PK complex. Together with previous structural and biochemical studies, our structures lead to a ...Cryo-Em Structure Of The... · Sign Up For Pnas Alerts · Results
  17. [17]
    Autophosphorylation and Self-Activation of DNA-Dependent Protein ...
    Jul 19, 2021 · It is known that DNA-PKcs undergoes autophosphorylation and phosphorylates other NHEJ factors including Ku70, Ku80, Artemis, PNKP and XRCC4 and ...
  18. [18]
    [PDF] Optimization of a LanthaScreen Kinase assay for DNA-PK
    Using the ATP Km,app concentration of ATP determined in step 1, a kinase titration is performed in order to determine the amount of kinase required to elicit an ...Missing: kinetic | Show results with:kinetic
  19. [19]
    Secrets of DNA-PKcs beyond DNA repair | npj Precision Oncology
    Jul 23, 2024 · ... DNA-dependent protein kinase catalytic subunit (DNA-PKcs). In 1985, Walker et al. first demonstrated that the addition of double-stranded ...
  20. [20]
    Structural insights into inhibitor regulation of the DNA repair protein ...
    Jan 5, 2022 · The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) has a central role in non-homologous end joining, one of the two main pathways
  21. [21]
    LRIK interacts with the Ku70–Ku80 heterodimer enhancing the ... - NIH
    Jun 25, 2020 · ... (DNA-PKcs), which binds Ku separately. DNA-PKcs recruitment stabilizes Ku complex binding at the DSB and is the protein kinase responsible ...
  22. [22]
    DNA end resection and its role in DNA replication and DSB repair ...
    Oct 30, 2020 · Sufficient end resection is important for RPA complex and RAD51 loading, which are essential for homologous recombination and error-free repair.Extension Of Dna End... · Termination Of Dna End... · Fig. 3. Dna End Resection...
  23. [23]
  24. [24]
    DNA-dependent protein kinase suppresses double-strand break ...
    DNA-PKcs may suppress spontaneous HR through NHEJ of spontaneous DSBs, perhaps at stalled or blocked replication forks.
  25. [25]
    DNA-PKcs suppresses illegitimate chromosome rearrangements
    We found that DNA-PKcs regulates opposing DNA end resection ... homologous end joining in G1 but is distinct from resection during homologous recombination.
  26. [26]
    Repair of G1 induced DNA double-strand breaks in S-G2/M ... - Nature
    Oct 16, 2020 · The alternative non-homologous end-joining (NHEJ) pathway promotes DNA double-strand break (DSB) repair in cells deficient for NHEJ or homologous recombination.
  27. [27]
    DNA double strand break repair via non-homologous end-joining
    As stated above, recruitment of DNA-PKcs to the DNA-Ku complex results in translocation of the Ku heterodimer inward on the DNA, likely to allow DNA-PKcs access ...
  28. [28]
    DNA-dependent protein kinase in nonhomologous end joining
    Conclusions and speculations. It is becoming increasingly clear that DNA-PKCS plays a central role during the NHEJ process. This enzyme not only captures and ...
  29. [29]
    Coordination of DNA–PK Activation and Nuclease Processing of ...
    Nonhomologous end joining (NHEJ) is a major pathway responsible for the repair of these complex breaks. Proteins involved in NHEJ include the Ku 70/80 ...Dna Double-Strand Breaks · Repairing Dna Dsbs · Dna--Pk Structure And...
  30. [30]
    Comparison of nonhomologous end joining and homologous ... - NIH
    Since the majority of randomly occurring DSBs have DNA ends that require processing prior to ligation we conclude that in proliferating cells NHEJ repairs 75% ...
  31. [31]
    The multifaceted functions of DNA‐PKcs: implications for the therapy ...
    Jun 19, 2024 · DNA‐PKcs can be trans‐phosphorylated or auto‐phosphorylated on at least 40 sites located in different phosphorylation clusters such as JK, PQR, ...<|control11|><|separator|>
  32. [32]
    Genome editing with the HDR-enhancing DNA-PKcs inhibitor ...
    Nov 27, 2024 · DNA-PKcs plays a crucial role in both promoting NHEJ and repressing HDR, and inhibiting DNA-PKcs increases HDR across multiple loci and cell ...
  33. [33]
    V(D)J Recombination: Recent Insights in Formation of the ...
    Apr 29, 2022 · V(D)J recombination is an essential mechanism of the adaptive immune system, producing a diverse set of antigen receptors in developing lymphocytes.
  34. [34]
    Overview of methodologies for T-cell receptor repertoire analysis
    Jul 10, 2017 · VDJ recombination of the different TCR genes could theoretically generate between 1015 and 1020 TCR chains. Despite this, the actual diversity ...
  35. [35]
    Three-dimensional structure of the human DNA-PKcs/Ku70/Ku80 ...
    Dimeric particles are observed in which two DNA-PKcs/Ku70/Ku80 holoenzymes interact through the N-terminal HEAT repeats. The proximity of the dimer contacts ...
  36. [36]
    Cryo-EM structure of human DNA-PK holoenzyme | Cell Research
    Aug 25, 2017 · DNA-dependent protein kinase (DNA-PK) is a serine/threonine protein kinase complex composed of a catalytic subunit (DNA-PKcs) and KU70/80 ...Missing: composition | Show results with:composition
  37. [37]
    Defining interactions between DNA-PK and ligase IV/XRCC4
    DNA-PK and ligase IV/XRCC4 interact at DNA termini which results in stimulated ligase activity. Here, we define interactions between the components of these two ...Missing: partners | Show results with:partners
  38. [38]
    Hsp90 Inhibitor STA9090 Sensitizes Hepatocellular Carcinoma to ...
    Aug 10, 2021 · Our results suggest that Hsp90 inhibitor STA9090 decreases DNA-PKcs protein stability and PRKDC mRNA level, which provide a theoretical basis for the promising ...
  39. [39]
    DNA-PKCS binding to p53 on the p21WAF1/CIP1 promoter blocks ...
    Inhibition of DNA-PK activity prevents DNA-PKCS binding to p53 on the p21 promoter, restores p21 transcription and significantly reduces cell death.
  40. [40]
    A Novel Role for DNA-PK in Metabolism by Regulating Glycolysis in ...
    Apr 1, 2022 · DNA-PK impacts glycolysis through direct interaction with glycolytic enzymes and modulation of enzymatic activity.
  41. [41]
    DNA-dependent protein kinase: DNA binding and activation ... - PNAS
    Because the DNA binding of p460 is salt-labile and competed by supercoiled plasmid DNA, one role for Ku may be to stabilize the binding of p460 to DNA ends.
  42. [42]
    Autophosphorylation of DNA-PK CS regulates its dynamics at DNA ...
    Apr 16, 2007 · Many in vitro studies have shown that the Ku70/80 heterodimer directs DNA-PKCS to DNA ends and stabilizes the DNA–DNA-PKCS interaction (Smith ...
  43. [43]
    Mapping of protein-protein interactions within the DNA-dependent ...
    These data suggest that Ku has evolved to become part of the DNA-PK holo-enzyme by acquisition of a protein-protein interaction motif at the C-terminus of Ku80.Results · Ku80 Interacts With Dna-Pkcs · The Ku80 C-Terminal Region...Missing: labs composition
  44. [44]
    DNA-PKcs-interacting protein KIP binding to TRF2 is required for the ...
    DNA-PKcs-interacting protein KIP interacts with TRF2 and enhances the telomere binding activity of TRF2. Depletion of KIP induces telomere-damage response.Yeast Two-Hybrid Screening · Gst Pull-Down... · Ubiquitination Assay
  45. [45]
    The multifaceted functions of DNA‐PKcs: implications for the therapy ...
    Jun 19, 2024 · DNA-PKcs is involved in nervous system diseases, senescence, asthma, cardiovascular disease, cancers, chronic kidney diseases, ...Missing: parameters | Show results with:parameters
  46. [46]
    DNA-PKcs: A Targetable Protumorigenic Protein Kinase
    DNA-PKcs is a pleiotropic protein kinase that plays critical roles in cellular processes fundamental to cancer.Missing: timeline | Show results with:timeline
  47. [47]
    The plié by DNA-PK: dancing on DNA - ScienceDirect.com
    Feb 18, 2021 · Activated DNA-PKcs is auto-phosphorylated at two well-characterized clusters, the PQR cluster (aa 2023-2056) and the ABCDE cluster (aa 2609-2647) ...
  48. [48]
    Unraveling the Complexities of DNA-Dependent Protein Kinase ...
    DNA-dependent protein kinase (DNA-PK) orchestrates DNA repair by regulating access to breaks through autophosphorylations within two clusters of sites (ABCDE ...
  49. [49]
    Phosphorylation in the serine/threonine 2609–2647 cluster ...
    End joining promoted by mutant DNA-PKcs proteins is still dependent on DNA-PK-catalyzed phosphorylation. The finding that the D6 mutant promotes end joining ...Missing: phosphomimetic T2609E
  50. [50]
    Catalytic subunit of DNA-dependent protein kinase - NIH
    The striking similarity between DNA-PKcs−/− and SCID mice in terms of their lymphocyte development and V(D)J recombination suggests that the “leaky” phenotype ...
  51. [51]
    Targeted Disruption of the Catalytic Subunit of the DNA-PK Gene in ...
    Although our DNA-PKcs defective mice closely resemble Scid mice, they differ by having elevated numbers of CD4+CD8+ thymocytes. This suggests that the Scid mice ...
  52. [52]
    V(D)J recombination activates a p53-dependent DNA damage ...
    "Broken" V(D)J coding ends accumulate in scid lymphocyte precursors as a consequence of a mutation in DNA-dependent protein kinase (DNA-PK). The ensuing ...
  53. [53]
    DNA-PKcs Is Involved in Ig Class Switch Recombination in Human B ...
    Dec 15, 2015 · In this study, we examined the pattern of recombination junctions from in vivo-switched B cells from two DNA-PKcs-deficient patients.
  54. [54]
    Kinase-dependent structural role of DNA-PKcs during ... - PNAS
    Aug 2, 2018 · Together, our findings identify a kinase-dependent role of DNA-PKcs in suppressing MH-mediated end joining and a structural role of DNA-PKcs ...
  55. [55]
    IgH Class Switch Recombination to IgG1 in DNA-PKcs-Deficient B ...
    To assess the role of the DNA-PKcs nonhomologous DNA end-joining (NHEJ) protein in Ig heavy chain class switch recombination (CSR), we assayed CSR ability ...
  56. [56]
    Elevated DNA double strand breaks and apoptosis in the CNS of ...
    Apr 23, 2001 · ... DNA-PKcs knock-out mice did not find CNS defects. This apparent discrepancy may be technical since the previous study relied on pyknotic ...
  57. [57]
    DNA-PK promotes the survival of young neurons in the embryonic ...
    May 7, 2010 · Remarkably, diminished DNA-PK activity resulted in selective death of recently born neurons, an observation corroborated in the embryonic retina ...
  58. [58]
    Defective embryonic neurogenesis in Ku-deficient but not DNA ...
    Herein, we show that Ku70 and Ku80 deficiency but not DNA-PKcs deficiency results in dramatically increased death of developing embryonic neurons in mice. The ...Missing: knockout | Show results with:knockout
  59. [59]
    DNA-PK inhibition sustains the antitumor innate immune ... - NIH
    Feb 1, 2025 · Among the innate immune pathways that may be triggered by cytosolic dsDNA and RNA sensors activation induced by anticancer therapies, we ...
  60. [60]
    The crosstalk between DNA-damage responses and innate immunity
    Oct 25, 2024 · DNA-PK is involved in IFN signaling pathways to induce the expression of IFNs and inflammatory cytokines in various types of cells. It is ...
  61. [61]
    Article DNA-PKcs is required for cGAS/STING-dependent viral DNA ...
    Jan 19, 2024 · We show that DNA-PK is essential for cyclic GMP-AMP synthase (cGAS)- and stimulator of interferon genes (STING)-dependent IFN-I responses in human cells.<|control11|><|separator|>
  62. [62]
    TRF2/RAP1 and DNA–PK mediate a double protection against ...
    ... TRF2/RAP1 and DNA–PK complexes protect telomeres by two complementary mechanisms. The former prevents C-NHEJ-mediated end fusion at the initial DNA–PK end ...
  63. [63]
    Chromosome end protection by RAP1-mediated inhibition of DNA-PK
    Apr 16, 2025 · Here we show that the shelterin components TRF2 and RAP1 form a complex with DNA-PK that directly represses its end-joining function at telomeres.
  64. [64]
    [PDF] Chromosome end protection by RAP1-mediated inhibition of DNA-PK
    Dec 28, 2024 · Cryo-EM structure of the RAP1:DNA-PK complex. a. Composite electron. 686 density map with protein domains coloured as indicated. Schematic ...<|control11|><|separator|>
  65. [65]
    Microhomology-mediated end joining: a back-up survival ...
    Microhomology-mediated end joining (MMEJ) is an error-prone repair mechanism that involves alignment of microhomologous sequences internal to the broken ends ...
  66. [66]
    Microhomology-mediated End Joining and Homologous ... - PNAS
    Microhomology-mediated end joining (MMEJ) is a major pathway for Ku-independent alternative nonhomologous end joining, which contributes to chromosomal ...
  67. [67]
    DNA-PKcs is critical for telomere capping - PMC - NIH
    The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is critical for DNA repair via the nonhomologous end joining pathway.Results And Discussion · Figure 3 · Table 3Missing: conditional dysfunction
  68. [68]
    Spontaneous Tumor Development in Bone Marrow Rescued DNA ...
    An increase in telomere fusions was previously reported in DNA-PKcs knockout mouse cells as well as in the recently described DNA-PKcs kinase dead mutant mouse ...Results · Figure 2. Dna-Pkcs... · Telomere Dysfunction In...Missing: aneuploidy | Show results with:aneuploidy
  69. [69]
    Harnessing the cooperation between DNA‐PK and cGAS in cancer ...
    May 5, 2023 · Genetic alterations of PRKDC, the gene encoding DNA-PKcs, are found in numerous cancer types with a frequency above 10%. Several somatic ...Missing: percentage | Show results with:percentage
  70. [70]
    Secrets of DNA-PKcs beyond DNA repair - PMC - PubMed Central
    Jul 23, 2024 · This review mainly discusses these non-canonical roles of DNA-PKcs in cellular biology and their potential contribution to tumorigenesis.Missing: alternative | Show results with:alternative
  71. [71]
    Impact of PARP-1 and DNA-PK expression on survival in ... - PubMed
    Conclusion: This hypothesis generating study showed that high tumor levels of DNA-PK correlate with poor survival of GBM patients.Missing: 1.5-2.0 | Show results with:1.5-2.0
  72. [72]
    DNA-PKcs promotes fork reversal and chemoresistance
    Sep 20, 2022 · DNA- PKcs inhibition in BRCA1/2-deficient cancer cells with acquired fork stability prevents fork slowing and restores cellular sensitivity to ...
  73. [73]
    A DNA-PKcs mutation in a radiosensitive T-B- SCID patient inhibits ...
    Here we have identified the first human RS-SCID patient to our knowledge with a DNA-PKcs missense mutation (L3062R). The causative mutation did not affect ...
  74. [74]
    PRKDC mutations in a SCID patient with profound neurological ... - JCI
    Jun 3, 2013 · In mice, loss of DNA ligase IV or XRCC4, both essential NHEJ proteins, is embryonic lethal, with marked apoptosis in the embryonic neuronal ...<|control11|><|separator|>
  75. [75]
  76. [76]
    Severe Combined Immunodeficiency (SCID) - Medscape Reference
    Nov 30, 2023 · The accurate incidence of SCID in the United States is unknown, but it has been estimated to be in 1 per 50,000-100,000 births across all ethnic ...Practice Essentials · Pathophysiology · Epidemiology
  77. [77]
    Generation and Characterization of Severe Combined ...
    Sep 27, 2012 · SCID mice, which arose spontaneously due to the defective DNA-PKcs gene (Prkdc), show an immunodeficient phenotype and increased sensitivity ...Resource · Results · Generation Of Scid Rats...Missing: traits | Show results with:traits
  78. [78]
    Impaired synaptic transmission and long-term potentiation...
    Apr 2, 2025 · We also showed that DNA-PKcs knockout mice, lacking both protein and enzymatic activity, present a deficit in long-term potentiation (LTP) along ...
  79. [79]
    Do DNA double-strand breaks drive aging? - PMC
    DSBs trigger the DNA damage response (DDR) that directs a cell to repair the break, undergo apoptosis or become senescent. There is increasing evidence that the ...Missing: exhaustion | Show results with:exhaustion
  80. [80]
    DNA–dependent protein kinase in telomere maintenance and ...
    Jan 17, 2020 · The catalytic DNA–PKcs is able to interact with TRF2 and RAP1 at telomeres, and their association prevents end–joining [17]. Additionally, the ...
  81. [81]
    The central role of DNA damage in immunosenescence - Frontiers
    Uncapping of eroded telomeres is sensed as a DSB, triggering a persistent DDR and permanent exit from the cell cycle. The shortening of telomeres towards this ...
  82. [82]
    Identification of a highly potent and selective DNA ... - PubMed
    NU7441, 8-dibenzothiophen-4-yl-2-morpholin-4-yl-chromen-4-one, is a highly potent and selective DNA-PK inhibitor with an IC50 of 14 nM.
  83. [83]
    NU7441 (KU-57788) | DNA-PK inhibitor | Mechanism | Concentration
    NU7441 (KU-57788) is a highly potent and selective DNA-PK inhibitor with IC50 of 14 nM and also inhibits mTOR and PI3K with IC50 of 1.7 μM and 5 μM in cell-free ...
  84. [84]
    Pharmacologic Inhibitor of DNA-PK, M3814, Potentiates ...
    May 4, 2020 · M3814 potently inhibits DNA-PK catalytic activity and sensitizes multiple cancer cell lines to ionizing radiation (IR) and DSB-inducing agents.
  85. [85]
    AZD7648 is a potent and selective DNA-PK inhibitor that enhances ...
    Nov 7, 2019 · We demonstrate that the potent and highly selective DNA-PK inhibitor, AZD7648, is an efficient sensitizer of radiation- and doxorubicin-induced DNA damage.Missing: IC50 | Show results with:IC50
  86. [86]
    7,9-dihydro-8H-purin-8-one (AZD7648), a Potent and Selective DNA ...
    Dec 18, 2019 · We screened our corporate collection for DNA-PKcs inhibitors with good PI3 kinase selectivity, identifying compound 1.
  87. [87]
    AZD7648 | DNA-PK inhibitor | Mechanism - Selleck Chemicals
    AZD7648 is a potent inhibitor of DNA-PK with an IC50 of 0.6 nM in biochemical assay and more than 100-fold selective against 396 other kinases.<|control11|><|separator|>
  88. [88]
    A first-in-man phase 1 study of the DNA-dependent protein kinase ...
    Nov 24, 2020 · Pharmacological inhibitor of DNA-PK, M3814, potentiates radiotherapy and regresses human tumors in mouse models. Mol. Cancer Ther. 19, 1091–1101 ...
  89. [89]
    Study Details | NCT04555577 | Peposertib and Radiation Therapy ...
    I. To evaluate pharmacodynamic properties of M3814. II. To assess the alterations in tumor immune microenvironment as a result of deoxyribonucleic acid (DNA)- ...Missing: 2024 | Show results with:2024
  90. [90]
    CTNI-42. PHASE I CLINICAL TRIAL OF PEPOSERTIB PLUS ...
    We conducted a two-stage phase I trial of peposertib plus radiation in newly-diagnosed MGMT-unmethylated glioblastoma (NCT04555577). ... RESULTS. Fifteen ...Missing: ORR 2024
  91. [91]
    Study Details | NCT04092270 | ClinicalTrials.gov - Clinical Trials
    This phase I trial studies the side effects and best dose of peposertib when given together with pegylated liposomal doxorubicin hydrochloride in treating ...
  92. [92]
    Targeting the DDR beyond PARP inhibitors: Novel agents and ...
    M3814/Nedisertib/Peposertib ... A phase I trial of KSQ-4279 as monotherapy is currently enrolling, with separate cohorts of combination therapy with PARP ...
  93. [93]
    DNA-PK inhibition enhances gene editing efficiency in HSPCs for ...
    Notably, a recently identified DNA-PKcs inhibitor with superior selectivity, AZD7648, has been shown to enhance HDR efficiency in vitro in cell lines, primary ...
  94. [94]
    Simultaneous inhibition of DNA-PK and Polϴ improves integration ...
    Aug 14, 2023 · AZD7648 treatment consistently reduced NHEJ-dependent InDels by up to 13.7-fold in HEK293T, 4.3-fold in Jurkat, 3-fold in HepG2, 6.5-fold in ...
  95. [95]
    Inhibition of DNA-PK with AZD7648 Sensitizes Tumor Cells to ...
    Here, we show that AZD7648 with radiotherapy not only increases cancer cell death via increased DNA damage, but also that cell death is immunogenic, inducing ...
  96. [96]
    DNA-PKcs Dysfunction Enhances the Antitumor Activity of ...
    Mar 19, 2025 · DNA-PK deficiency enhances cGAS-mediated antiviral innate immunity. Previous studies indicated that using the DNA-PK inhibitor Peposertib ...
  97. [97]
    The DNA-PK inhibitor AZD7648 alone or combined with pegylated ...
    May 17, 2025 · Neither anaemia nor myelosuppressive TEAEs were reported in a trial of single-agent peposertib [28], and trials of peposertib with other ...
  98. [98]
    The DNA Damaging Revolution: PARP Inhibitors and Beyond
    May 17, 2019 · PARP inhibitors are now approved for the treatment of ovarian cancer in different settings and BRCA-mutant breast cancers.
  99. [99]
    PRKDC: new biomarker and drug target for checkpoint blockade ...
    Apr 1, 2020 · The CT26 animal model was used to evaluate the role of PRKDC as a predictive biomarker and the efficacy of the DNA-PK inhibitor. Results.Missing: myelosuppression | Show results with:myelosuppression