Your privacy, your choice

We use essential cookies to make sure the site can function. We also use optional cookies for advertising, personalisation of content, usage analysis, and social media.

By accepting optional cookies, you consent to the processing of your personal data - including transfers to third parties. Some third parties are outside of the European Economic Area, with varying standards of data protection.

See our privacy policy for more information on the use of your personal data.

for further information and to change your choices.

Skip to main content

DNA-PKcs, a player winding and dancing with RNA metabolism and diseases

Abstract

The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a key kinase in the DNA repair process that responds to DNA damage caused by various factors and maintains genomic stability. However, DNA-PKcs is overexpressed in some solid tumors and is frequently associated with poor prognosis. DNA-PKcs was initially identified as a part of the transcription complex. In recent years, many studies have focused on its nonclassical functions, including transcriptional regulation, metabolism, innate immunity, and inflammatory response. Given the pleiotropic roles of DNA-PKcs in tumors, pharmacological inhibition of DNA-PK can exert antitumor effects and may serve as a potential target for tumor therapy in the future. This review summarizes several aspects of DNA-PKcs regulation of RNA metabolism, including its impact on transcriptional machinery, alternative splicing, and interaction with noncoding RNAs, and provides insights into DNA-PKcs beyond its DNA damage repair function.

Introduction

The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a serine/threonine protein kinase and a member of the phosphatidylinositol 3-kinase-related kinase (PIKK) family. DNA-PKcs has been broadly studied in the field of DNA damage repair, and it repairs broken DNA through nonhomologous DNA end-joining (NHEJ) to maintain genomic stability and integrity [1]. Briefly, the DNA-PK complex consists of Ku70, Ku80, and DNA-PKcs. When DNA double-strand breaks occur, Ku70/80 heterodimers recognize the DNA end and recruit DNA-PKcs to damaged sites, forming the DNA-PKcs-Ku-DNA complex [2]. Simultaneously, the serine/threonine protein kinase activity of DNA-PKcs is activated, which phosphorylates itself as well as a series of downstream proteins to repair damaged DNA. Dysregulation of DNA-PKcs expression and activity is usually tightly correlated with poor prognosis in patients with tumors. Therefore, DNA-PKcs are considered potential targets for tumor therapy [3]. Genetic and pharmacological methods to regulate DNA-PK function could affect downstream signal transduction and tumor proliferation as well as increase the radiosensitivity of tumor cells [4, 5].

Although the role of DNA-PKcs in DNA damage repair has been extensively studied, it was initially discovered as a part of the SP1 transcription complex [6]. The transcriptional regulation and gene expression status of cells changed when the inhibitor NU7441 was used. Subsequent studies revealed that DNA-PK plays an important role in transcriptional regulation. For example, DNA-PKcs alters the transcriptional activity and protein stability of some certain transcription factors and affects the transcription of downstream target genes. DNA-PKcs could also bind transcription factors, form transcription complexes, and regulate gene expression. In addition, some pathological factors, such as hypoxia and high NaCl concentration, can also induce transcriptional changes through DNA-PK [7, 8].

Compared with mRNA, noncoding RNAs such as long noncoding RNA (lncRNAs), micro RNA (miRNAs), and circular RNA (circRNAs) usually do not encode proteins. In recent years, researchers have found that noncoding RNA can affect the function of DNA-PKcs either by direct binding or by inhibiting its enzymatic activity. Moreover, DNA-PKcs influences ribosomogenesis as well as rRNA synthesis and translation. In addition to affecting mRNA transcription, DNA-PKcs also affects the alternative splicing of mRNA, which in turn affects the epigenetic regulation of genes, including SRSF1 and SRSF2 [9].

In this review, we provide a detailed summary of DNA-PK functions in RNA metabolism, including transcriptional regulation (Fig. 1), noncoding RNA, and alternative splicing.

Fig. 1
figure 1

The pleiotropy of DNA-PKcs in transcriptional regulation. DNA-PKcs could affect gene transcription processes from different dimensions. It participates in basic transcription programs including transcription initiation, activation, and elongation. For some certain transcription factors, DNA-PKcs regulates transcriptional activity and protein stability of transcription factors to affect the function. In several transcription complexes, DNA-PKcs functions as an indispensable component. Moreover, DNA-PKcs is also involved in transcription of fatty acid synthase genes and regulating the differentiation of immune cells and the transcription of cytokine. In addition, DNA-PKcs could regulate DDR-related genes transcription processes in response to DNA damage. The figure was created with BioRender.com. P, phosphorylation; Ub, ubiquitination

DNA-PKcs in transcriptional regulation

DNA-PKcs was initially found as a part of transcriptional complex. Subsequently, researchers investigated global gene transcription changes in mammalian cells with DNA-PKcs inhibition or deficiency. It is reported that in vitro transcription levels of DNA-PK-deficient Chinese hamster ovary (CHO) cell extracts were reduced by twofold to sevenfold compared with the control cells [10]. Biological processes, including transcription and regulation of gene expression, changed after depletion of DNA-PKcs or treatment with inhibitor NU7441 [11]. Similarly, according to a recent study, the transcriptome changed after the treatment of NU7441 in human BT549 triple-negative breast cancer cells. RNA-seq results showed that there were significant differences in the genes of the pattern recognition receptor and cytoplasmic DNA-sensing pathways [12]. Although this indicates that DNA-PKcs may regulate global gene transcription, the mechanism has not been thoroughly explored.

The functions of DNA-PKcs on the transcription of certain genes including laminin alpha 4 (Lama4), NF-E2-related factor 2 (Nrf2), and human xanthine oxidoreductase gene (hXOR) were reported. Initially, F Bryntesson et al. found that Lama4 gene transcription was regulated by DNA-PK through a radiation-independent manner in primary mouse embryo fibroblasts [13]. Nrf2 is upregulated in various types of cancer and identified as a pharmacological target to overcome chemotherapy resistance. Wogonin could reverse resistance by inhibiting Nrf2/ARE pathway. DNA-PKcs are involved in wogonin-induced downregulation of Nrf2 mRNA at the transcriptional level in human myelogenous leukemia K562/A02 cells [14]. DNA-PKcs contributes to the regulation of hXOR mRNA expression induced by oncostatin M (OSM) [15]. Interestingly, DNA-PK could inhibit myelin transcription factor 1-like (MYT1L) mediated transcription in p53 mutated glioblastoma cells [16]. DNA-PK regulates Wnt signal transduction to control the metastatic phenotype in castration-resistant prostate cancer (CRPC) cells by interacting with the transcription factor LEF1. However, the molecular mechanism needs to be clearly elucidated [17].

It is mysterious to investigate how DNA-PKcs selectively regulate certain gene transcription or the global gene transcription.

DNA-PKcs in transcription initiation, activation/repression, and elongation

Canonically, the global gene transcription program involves transcription initiation, activation, and elongation. DNA-PKcs acts as an additional component of the DNA topoisomerase IIβ (TopoIIβ) and poly[adenosine diphosphate (ADP)–ribose] polymerase–1 (PARP-1) complex to mediate transcription initiation [18, 19]. Consistently, other studies reported the presence of DNA-PKcs on the RNA polymerase II (Pol II) transcription units. DNA-PKcs is crucial for Pol II extension during transcriptional activation. Pause release, elongation of Pol II, and γH2AX levels were relatively reduced after using DNA-PKcs inhibitors [20, 21]. Furthermore, DNA-PK forms a complex with WW domain-containing protein 2 (WWP2) and RNA Pol II in response to DNA double-strand break (DSB). Proteasomes were recruited directly, leading to the degradation of RNA Pol II on damaged chromatin, thereby silencing the transcription of damaged genes [22]. DNA-PKcs can also remove RNA Pol II from DNA to induce transcriptional arrest in response to DSB [23]. Moreover, DNA-PKcs could prevent Ku heterodimers from overloading at a single DNA end, physically protecting transcription in the vicinity of DNA ends [24]. These findings suggest that DNA-PKcs may participate in transcription in the vicinity of DNA break sites.

Under most circumstances, DNA-PK regulates transcriptional activity of transcription factors through phosphorylation. Heat shock transcription factor 1 (HSF1) interacts with DNA-PK, and purified HSF1 stimulates DNA-PK phosphorylation in vitro. DNA-PK may also maintain the activated form of HSF1, thereby prolonging its transcriptional activity [25]. In addition, DNA-PK could phosphorylate the progesterone receptor (PR) in a DNA-dependent manner, and the inhibition of DNA-PK reduces the activation of RNA Pol II and the transcriptional activity of PR [26]. Moreover, octamer transcription factor-1 (Oct-1) was regarded as a stress sensor that promotes cell survival subsequent to DNA damage. DNA-PK could regulate the activity of the Oct-1 in response to DSB [27, 28]. However, the mechanism of action of the Q-rich domain of Oct-1 during this process is still unclear.

Hypoxia increases the stability and activity of the hypoxia-inducible factors (HIFs). DNA-PK assembles with hypoxia-inducible factor 1 (HIF-1) and tripartite motif containing protein 28 (TRIM28) to form heterotrimers, which stabilize the HIF occupancy of hypoxia response elements (HREs) and recruit cyclin-dependent kinase 9 (CDK9) to phosphorylate Pol II C-terminal domain (CTD) on Ser2, leading to HIF transcriptional activation in hypoxic breast cancer cells [7].

DNA-PK interacts with forkhead box A2 (FoxA2) and promotes its transcriptional activation by phosphorylating its Ser283 site [29]. It is recruited to the sites of AR action and initiates p300 occupancy, facilitating AR-dependent transcriptional transactivation [11].

DNA-PK affects transcription mediated by the Ets transcription factor family, which includes Ets-1, ERG, and ELF5. Ets-1 activates gene transcription involved in various cellular pathways, including proliferation, differentiation, adhesion, migration, and invasion. Its overexpression is associated with inflammation, invasive lesions, and cancer. The DNA-PK complex, as a partner of Ets-1, phosphorylates Ets-1 and regulates its transcriptional activity in a DNA-independent manner [30, 31]. Additionally, DNA-PKcs is a regulatory factor of ERG transcriptional activity, and ERG binds to DNA-PKcs at the Tyr373 site, inhibiting DNA-PKcs and altering the transcriptional activity of ETS target genes [32]. Moreover, the interaction between ELF5 and DNA-PKcs inhibits the activity of ELF5, thereby affecting partial transcription [33].

Zinc finger protein of cerebellum 2 (Zic2) is a transcription activator that plays a crucial role in forebrain development in mammals. The molecular mechanism of Zic regulation of transcription is mainly related to two types of complexes containing Zic2. As a component of the complex I, DNA-PKcs could phosphorylate Zic2, which is a necessary step for the transformation of complex I to complex II (Fig. 2a). It is also important for the interaction and subnuclear co-localization of Zic2 and RNA helicase A (RHA). Zic2 activates the transcription of target genes by binding to DNA and recruiting RHA. Transcriptional activation of Zic2 also depends on DNA-PK in vivo. Researchers found that Zic2 Ser200 may be the main phosphorylation site of DNA-PKcs. The interaction between Zic2 and RHA is reduced and transcriptional activation was significantly reduced when the mutation occurred [34, 35]. However, the factors affecting the disruption of complex II and the dephosphorylation of Zic2 are still unknown.

Fig. 2
figure 2

The mechanism of transcriptional regulation by DNA-PKcs. a DNA-PKcs could phosphorylate Zic2, and regulate the transformation of complex I to complex II. b DNA-PKcs induces mtDNA fragmentation and inhibits mitochondrial open-reading-frame of the 12S rRNA type-C (MOTS-c) transcription. c DNA-PKcs is an upstream regulator of SOX2 to stabilize the SOX2 protein and promote glioma stem cell maintenance. d DNA-PKcs phosphorylates Egr1, thereby promoting IL2 expression. e DNA-PKcs phosphorylates NEMO, promotes its nuclear translocation, and triggers NF-κB activation. f DNA-PKcs phosphorylates YB-1, promoting its nuclear translocation and enhancing DNA repair. The figure was created with BioRender.com. P, phosphorylation; Ub, ubiquitination

In insulin signal pathway, DNA-PK phosphorylates the Ser262 site of upstream stimulatory factor 1(USF-1) and then binds to the promoter region of fatty acid synthase (FAS), mediating the transcriptional activation of lipogenesis under feeding/insulin treatment [36]. USF is necessary for regulating FAS promoter activity during fasting/feeding. This study elucidated the relationship between DNA-PKcs and the insulin signal pathway. However, the interaction between transcriptional activation during the feeding process and the component proteins involved in DDR in the promoter region of adipogenic genes requires further exploration.

Cryptochrome (CRY) is a key component of the transcriptional repression complex and is involved in the regulation of the mammalian circadian clock. DNA-PK may indirectly and negatively regulate the phosphorylation of Ser588 at the C-terminus of CRY1. Loss or inhibition of DNA-PK leads to long-period rhythms [37].

DNA-PK could phosphorylate Ser435 and Ser446 of human serum response factor (SRF) [38]. The c-Jun Ser249 site is phosphorylated by DNA-PK in vitro, but the phosphorylation does not interfere with DNA binding [39].

In addition to affecting the phosphorylation of transcription factors, DNA-PKcs is also indispensable in several transcription complexes. The MEGATrans complex is necessary for activating enhancer RNA transcription and recruiting coactivators. It serves as a platform for recruiting specific enzymes (such as DNA-PK) to exert their effects. It serves as a hallmark that distinguishes the most active enhancers of the estrogen-regulated transcriptional program. Knockdown of DNA-PKcs or treatment with NU7441 significantly inhibited the activation of the estrogen receptor α-binding enhancer and its target genes expression [40]. The transcription factor tonicity-responsive enhancer/osmotic response element-binding protein (TonEBP/OREBP) can be activated by high NaCl concentrations, thereby increasing the transcription of protective genes. The DNA-PK subunit exists together with TonEBP/OREBP in the protein complexes formed in vitro. However, it is not clear whether it belongs to a part of the intracellular complex [8]. Moreover, DNA-PKcs interacts with other complexes, including transcriptional activator p300, myogenic determination factor MyoD, Akt1, and Akt2, to activate the transcription of the differentiation factor myogenin at its promoter without inducing DNA damage and repair [41].

Interestingly, DNA-PKcs induces mtDNA fragmentation through pathological mitochondrial division, inhibits MOTS-c transcription, and causes cardiac microcirculation dysfunction [42] (Fig. 2b). This suggests that DNA-PKcs may play a greater role in the transcriptional regulation of organelle genes.

DNA-PKcs in regulation of transcription factors protein stability

In addition to the impact on transcriptional activity, previous studies have also investigated the role of DNA-PK in the stability of transcription factor proteins. DNA-PK phosphorylates pancreatic duodenal homeobox-1 protein (PDX-1) at the Thr11 site in response to DNA damage in mouse pancreatic beta cells. Moreover, PDX-1 is further degraded by proteasomes to reduce the expression of GLUT2 and glucokinase genes [43]. Hypoxia mainly induces changes in the higher-order chromatin structure, initiating DNA-PK activation in the absence of DNA lesions. Furthermore, DNA-PK is activated and protects HIF-1α from degradation, contributing to the expression of GLUT1 and adaptation to hypoxia [44]. However, DNA DSB also partially contributes to DNA-PK activation.

Moreover, DNA-PK promotes the malignant phenotype of tumors. Snail1 is a key regulator of epithelial-mesenchymal transition (EMT). The direct interaction between DNA-PKcs and Snail1 induces Snail1 phosphorylation at Ser100, increasing the stability of the Snail1 protein and potentiating Snail1 functions. Ultimately, it promotes the invasive ability of the tumor [45]. However, phosphorylated Snail1 exhibits feedback inhibition of DNA-PK kinase activity, hindering DNA damage repair and ultimately inducing genomic instability [45]. This indicates the feedback mechanism of substrates on DNA-PKcs. Cellular myelocytomatosis oncogene (c-Myc) is dysregulated in various human cancers. DNA-PKcs regulates the phosphorylation level of c-Myc at Thr58 through the Akt/GSK3 β pathway in human cervix cancer HeLa cells. Overexpression of DNA-PKcs induces increased protein stability of c-Myc, thereby promoting cell proliferation and carcinogenic transformation [46]. Fang et al. reported that DNA-PK is an upstream regulator of SOX2, which is a core transcription factor that maintains the properties of stem cells. It phosphorylates the Ser251 of human SOX2 to prevent WWP2-mediated ubiquitination, thereby stabilizing the SOX2 protein and promoting glioma stem cell (GSC) maintenance under normal circumstances. However, DNA-PK is disassociated from SOX2, which facilitates SOX2 binding to WWP2 and promotes SOX2 ubiquitination and GSC differentiation in response to DNA damage [47] (Fig. 2c). This reveals the function of DNA-PK in maintaining GSCs by precisely regulating the stability of the SOX2 protein in the presence or absence of DNA damage.

DNA-PKcs in immune-related genes transcription

DNA-PKcs also participates in regulating the differentiation of immune cells and the transcription of cytokine. Research reported that DNA-PK regulated the differentiation of mouse CD4+ T cells into Th2 and Th1 cells by affecting the expression of GATA-3. Inhibition of DNA-PK through pharmacology or gene heterozygosity completely blocks the differentiation of CD4+ T cells into Th2 cells, as well as the expression of interleukin (IL)-5 and IL-13 [48]. DNA-PKcs plays a crucial role in the transcription of related genes after T cell receptor (TCR) stimulation. The Ser301 site of early growth response protein 1 (Egr1) is phosphorylated by DNA-PKcs in vitro. Inhibiting DNA-PKcs kinase activity reduces Egr1 protein expression by affecting protein stability, leading to a decrease in IL-2 level (Fig. 2d). Interestingly, phosphorylation of serine 301 of Egr1 is required for protein stability [49]. These studies provide critical targets for novel therapy for immune-related diseases. In addition, this also helps to understand the function of DNA-PKcs in immune cells.

DNA-PK acts as a DNA sensor upstream of the interferon (IFN) regulatory factor 3 (IRF-3) dependent innate immune response, triggering the transcription of type I interferon (IFN), cytokines, and chemokine genes [50]. When viral infection occurs, DNA-PK is activated to bind and phosphorylate the Thr135 site of IRF-3, reducing protein degradation caused by IRF-3 translocation to the cytoplasm and prolonging the half-life of IRF-3 [51].

The autoimmune regulatory factor (AIRE) is also regulated by DNA-PK. AIRE protein is a transcription factor expressed in myeloid thymic epithelial cells (mTECs) that plays a key role in central T cell tolerance. DNA-PK is involved in the phosphorylation of AIRE at Thr68 and Ser156, which influences its transactivation function [52]. In mice, DNA-PK is the direct molecular partner of AIRE, which cooperates with AIRE to induce peripheral tissue antigen transcription in mTECs [53]. In addition, mutated DNA-PKcs lead to impaired AIRE transcriptional activity in wild-type human fibroblasts, and DNA-PKcs may interact with AIRE to promote the expression of Toll-like receptor (TLR) 1, TLR3, and TLR8 in mouse RAW264.7 cells [54, 55].

Nuclear factor-kappa B (NF-κB) is a key transcription factor involved in immunity, inflammation, and cell transformation. Genetically toxic stress triggers phosphorylation of NF-κB essential modulator (NEMO) on the Ser43 site through the interaction between DNA-PK and NF-κB p65 subunit, allowing NEMO to shuttle through the nucleus and subsequently activate NF-κB (Fig. 2e). Moreover, DNA-PK could phosphorylate p50 subunit, promoting the stability of p50/p50 or p50/p65 dimers and their binding to promoters after tumor necrosis factor alpha treatment [56,57,58].

DNA-PKcs in DDR related genes transcription

DNA-PK regulates many non-DNA damage repair (DDR) related genes transcription processes. However, owing to extensive research on its classical functions, many studies have reported that DNA-PK regulates transcription to promote DDR response. For example, DNA-PK directly phosphorylates the transcriptional regulator Y-box binding protein (YB-1) at Thr89, resulting in nuclear translocation, enhanced activity, and increased cellular DNA repair level [59]. However, the mechanisms involved in the intracellular signal pathway after YB-1 nuclear translocation are still unclear (Fig. 2f). In addition, DNA-PKcs drives Bcl-2-associated transcription factor 1 (BCLAF1) to the nuclear membrane by directly phosphorylating the key BCLAF1 RS residues, thereby regulating DNA damage repair. The knockdown of BCLAF1 also leads to weakened binding between Ku70 and DNA-PKcs [60]. Moreover, phosphorylation of Krüppel-like factor 8 (KLF8) at the Ser80 site by DNA-PKcs is necessary for DNA damage [61]. Nuclear receptor subfamily 4 group A (NR4A) is an orphan member of the nuclear receptor family that can function as a ligand-independent transcription factor. It interacts with DNA-PKcs and is recruited to repair lesions after DNA damage. DNA-PK mediates NR4A phosphorylation, thereby regulating DSB repair in mammalian cells. Interestingly, NR4A transcriptional activity is not necessary [62].

To be investigated

Above all, the transcriptional regulation activity of DNA-PKcs largely depends on its kinase activity as the DNA-PK holoenzyme phosphorylation on other transcription machinery factors. The downstream mechanisms either involve conformational change or protein ubiquitination. However, the underlying mechanism remains unknown. It is necessary to use multi-omics approaches to identify the different binding/phosphorylated transcription factors for DNA-PK under different conditions, such as different organ development, disease progression, and subcellular organelle transcriptional regulation. Moreover, it is also important to discriminate the organization of DNA-PKcs/Ku70/Ku80 complex in DNA damage repair and transcription. Compared with the increasing resolution structures of DNA-PK complex in DNA damage repair, little is known about the DNA-PK transcription machinery. It is unknown how much Ku70/Ku80 contribute to the DNA-PKcs dependent transcription. Therefore, it is interesting to discriminate the Ku70/Ku80-dependent and Ku70/Ku80-independent roles in DNA-PKcs mediated transcription.

DNA-PKcs in noncoding RNAs interaction network

DNA-PK can interact with various noncoding RNAs, including long noncoding RNA (lncRNAs), micro RNA (miRNAs), circular RNA (circRNAs), and small Cajal body-associated RNA (scaRNAs).

LncRNAs

LncSNHG12 is a highly conserved lncRNA that can bind to DNA-PK, thereby promoting the ability of DNA-PKcs to bind to Ku70/Ku80 and regulate vascular DNA damage and cellular aging [63]. Lnc00312 directly binds to DNA-PKcs, obstructing the formation of the DNA-PK complex and inhibiting the NHEJ repair pathway, thereby enhancing the radiosensitivity of nasopharyngeal carcinoma cells [64]. Similarly, lncLINP1 and lncNEAT1 could also regulate DDR response through DNA-PKcs. LncLINP1 acts as a scaffold to interact with DNA-PKcs in response to DNA damage. Furthermore, knocking down LINP1 enhances the activation of radiation-induced apoptosis in HeLa cells [65]. LncNEAT1 is the core structural component of the nuclear paraspeckle (PS) organelles and it regulates DNA damage repair through ATM and DNA-PKcs [66]. In addition, HEXIM1 binds to lncNEAT1 to recruit the DNA-PK complex and paraspeckle proteins [67]. LncLINP1 binds to Ku80 and DNA-PKcs in the 5′ region (nucleotides 1–300) and 3′ region (nucleotides 600–917) of the transcript, enhancing DSB repair through the NHEJ pathway and promoting the resistance of triple-negative breast cancer cells to radiation and chemotherapy [68]. LncLEMGC interacts with DNA-PKcs and inhibits the phosphorylation of DNA-PKcs at Ser2056, ultimately inhibiting the progression and metastasis of gastric cancer by downregulating the ErbB1-SRC-FAK signaling pathway [69]. Research reported that methotrexate (MTX) activated DNA-PKcs in Jurkat cells and T cells, leading to the induction of TP53 and lncp21. Ultimately, lncp21 bound to RELA mRNA and inhibited NF-κB activity [70] (Fig. 3).

Fig. 3
figure 3

The interaction between DNA-PKcs and lncRNAs. The interaction between DNA-PKcs and lncRNA affects DNA repair, chemoradiotherapy resistance, and tumor progression. The figure was created with BioRender.com. P, phosphorylation

CircRNAs

CircUGGT2 combines with the KU heterodimer to recruit DNA-PKcs to damaged chromatin sites, thereby promoting H2AX phosphorylation and DSB repair [71]. CircRNA_001895 is elevated in sunitinib-resistant renal cell carcinoma (RCC). The silencing of CircRNA_001895 induced decreased levels of p-DNA-PK and Rad51 and increased levels of γH2AX [72]. This suggests a potential role for circRNAs in tumor drug resistance, but the mechanisms affecting DNA-PK phosphorylation need to be further investigated (Fig. 4a).

Fig. 4
figure 4

The interaction between DNA-PKcs and other noncoding RNAs. a The interaction between DNA-PKcs and circular RNAs. b ScaRNA2 weakens the interaction with Ku70/Ku80, thereby preventing the assembly and activation of the DNA-PK complex. c The roles of DNA-PKcs in rRNA processing and pre-rRNA biogenesis. d The interaction between DNA-PKcs and micro RNAs. The figure was created with BioRender.com. P, phosphorylation

MiRNAs

Unlike lncRNAs, most interactions between miRNAs and DNA-PKcs result in an impact on DDR repair in chemotherapy or radiotherapy. MiR-488-3p is significantly downregulated in clinical specimens and cell lines of malignant melanoma (MM) and its effect on the sensitivity of MM cells to cisplatin is mediated by DNA-PKcs. Overexpression of DNA-PKcs eliminates the promotion of miR-488-3p–induced sensitivity to cisplatin in MM cells [73]. MicroRNA-145 inhibits the translation of DNA-PKcs by binding to the 3′ untranslated region (UTR) of DNA-PKcs, resulting in accumulation of DNA damage [74]. Consistently, miR-1323 regulates the expression of DNA-PKcs protein by binding to the 3′-UTR of PRKDC, thereby participating in the radiation resistance of human lung cancer cells [75]. DSBs were induced by DNA-damage agents, then histone H2AX phosphorylation at Ser139 (γH2AX) increased. The level of γH2AX indicates the degree of DNA damage. Ectopic overexpression of miR-7 prolongs radiation-induced γH2AX lesion formation and reduces DNA-PKcs phosphorylation levels, thereby increasing the radiosensitivity of human cancer cells [76]. Similarly, ectopic overexpression of miR-101 effectively reduced ATM and DNA-PKcs levels in H1299, H1975, and A549 cells and increased radiosensitivity [77]. Moreover, research reported that melatonin upregulates DNA-PKcs by inhibiting miR-101 in HUVECs [78]. Genotoxic stress can also cause changes in miRNAs. Radiation induced transcription of miR-221 and miR-222. MiR-221 and miR-222 regulate DNA-PKcs expression by activating Akt, thereby affecting DNA damage repair [79]. These findings reveal the interaction network between noncoding RNA and DNA-PKcs (Fig. 4d).

ScaRNA

Besides lncRNAs, miRNAs, and circRNAs, DNA-PK could also interact with scaRNA2. After binding to DNA-PKcs, scaRNA2 weakens its interaction with Ku70/Ku80, thereby preventing the assembly and activation of the DNA-PK complex [80] (Fig. 4b).

Pre-rRNA biogenesis

In addition, DNA-PK plays an important role in ribosome biogenesis and may be a part of the pre-rRNA ribonucleoprotein complex. U3 snoRNA triggers Thr2609 site DNA-PKcs phosphorylation. Ku heterodimer mediates the catalytic deactivation or assembly of T2609 phosphorylation-defective DNA-PK, leading to rRNA processing defects [81].

Moreover, other researchers have found that DNA-PK may regulate pre-rRNA biogenesis in multiple steps, including pre-rRNA synthesis, modification, and processing. In addition, DNA-PK could recognize pre-rRNA. The absence of activated DNA-PK not only inhibits the transcription of 47S pre-rRNA but also suppresses the processing of the 18S rRNA precursor [82] (Fig. 4c).

DNA-PKcs in alternative splicing

Alternative splicing (AS) is an important mechanism of transcriptional regulation that generates transcriptome diversity. DNA-PKcs are involved in pre-mRNA alternative splicing. A set of pre‐mRNAs in human lung adenocarcinoma A549 cell cells treated with the topoisomerase II inhibitor MTX was altered including SRSF1, SRSF2, HNRNPDL, HNRNPH1, and PRPF38B [9]. This supports the involvement of DNA-PKcs in controlling pre-mRNA splicing in response to MTX-induced genotoxic stress.

In addition, researchers found that knockdown and pharmacological inhibition of DNA-PKcs resulted in significant negative enrichment of the cell “spliceosome” gene set, indicating interference of splicing activity in cells with impaired DNA-PKcs activity. Further research showed DNA-PKcs regulated RNA binding motif protein X-linked (RBMX) transcription by binding to its cis-regulatory elements proximal to the transcriptional start site. The DNA-PKcs-RBMX regulatory loop controls AR transcript maturation and downstream AR isoform transcriptional activity by affecting splicing decisions of appropriately spliced exons 2 and 3 [83].

Our previous study showed the global gene landscape of DNA-PKcs-RNA interactions in human osteosarcoma U2OS cell and KEGG pathway analysis showed that many RNAs were involved in cell adhesion and receptor ECM interaction pathways. In addition, we have demonstrated that DNA-PKcs affects CD44 alternative splicing, and the expression of CD44 variants increases after DNA-PK inhibition [84]. This provides insights into how alternative splicing can promote DNA-PK mediated DDR responses.

Perspectives and conclusions

Although many studies have reported the non-DDR function of DNA-PKcs, there are still several molecular mechanisms to be explored [85].

The intersection of classical and nonclassical functions of DNA-PKcs

As mentioned earlier, DNA-PK is involved in the biogenesis of rRNA without genotoxic stress. Simultaneous treatment of low-dose DNA-PK-i and PARP-i will induce synthetic lethality in BRCA1 deficient triple negative breast cancer cell lines, which is caused by defects in pre-rRNA biogenesis rather than DNA damage reaction. Interestingly, DNA-PK exhibited the opposite function after genotoxic stress. DNA-PK acts upstream of PARP-1 and inhibits rRNA synthesis after DNA damage. DNA-PK and PARP-1 inhibitors prevented cisplatin induced rRNA synthesis blockade [86].

The nucleolus is located in the core of the nucleus and occupies a large amount of space in the nucleus. Its main function is to produce and assemble ribosome complexes [87, 88]. Gamma H2AX is a substitute marker for DSB and a substrate for DNA-PKcs, mainly present in the nucleoplasm and not in the nucleolus. Studies have found that DNA-PKcs exists in the nucleolus even in the absence of genetic stress toxicity [82]. This indicates a correlation between the subcellular localization and function of DNA-PKcs. However, the regulatory mechanism is not clear. Ku heterodimer and pre-rRNA may have some functions; it is worth further exploration.

DNA-PKcs in the intersection of DNA damage repair and transcription

DNA lesions from both exogenous and endogenous-derived insults will directly or indirectly impede the transcription. The transcription-coupled DNA repair (TCR) pathway is crucial to eliminate such DNA lesions. When DSB occurs within or near actively transcribed genes, DNA-PK and HECT E3 ubiquitin ligase WWP2 are recruited to the damage sites, leading to ubiquitination of RNAPII subunit RPB1 and triggering RNAPII degradation, thereby promoting transcriptional silencing of broken genes [22]. However, it is unknown if DNA-PKcs functions in the TCR machinery or in the repair of transcription-induced DNA breaks or in the resolution of transcription-replication conflicts (TRCs) [89].

Ku70/Ku80 cooperation with DNA-PKcs in DDR and RNA metabolism

Ku70/Ku80, as a part of the DNA-PK complex, also plays an important role in DDR and RNA metabolism. Ku70/Ku80 recognizes and binds to the end of DNA, and the kinase activity of DNA-PKcs is fully activated only in the presence of DNA and Ku70/Ku80 [90, 91]. The N-terminal α-solenoid of DNA-PKcs consists of an N-terminal and middle Huntingtin, elongation factor 3, PP2A, and TOR1 (HEAT) repeats (N-HEAT and M-HEAT) [92]. N-HEAT is divided into two fragments (NH1 and NH2), while M-HEAT is divided into three fragments (MH1, MH2, and MH3). The cryoelectron microscopy structures of DNA-PK reveals that Ku stabilizes the DNA-PKcs DNA complex by prolonging DNA binding, causing DNA-PKcs to switch to an activated state. Moreover, Ku contacts NH1 of N-HEAT and MH3 of M-HEAT, bridging both sides of the DNA binding slot in DNA-PKcs and fixing N-HEAT of DNA-PKcs onto DNA [93]. Ku binds to substrates to promote DNA-PKcs phosphorylation of substrates. In vitro experiments have shown that the binding of Oct-1 to Ku promotes DNA-PK phosphorylation of Oct-1 [94]. In addition, the Ku protein can also affect the nonclassical functions of DNA-PKcs. Ku drives the assembly of DNA-PKcs on RNA, promoting the biogenesis of ribosomes. Interestingly, Li et al. also demonstrated that DNA-PK regulates rRNA biogenesis by phosphorylating ribosomal proteins. In addition, they found that pre-rRNA is a functional partner of the Ku70/Ku80 complex in the nucleolus. In addition, the Ku subunit is necessary for targeting DNA-PKcs to appropriate positions in the nucleolus. Therefore, they proposed a positive feedback hypothesis between Ku70/Ku80 complex, pre-rRNA, and DNA-PKcs. The interaction between Ku complex and pre-rRNA may mediate the activation of DNA-PKcs in the nucleolus [82].

The prospects of DNA-PK targeting based therapy

Numerous studies have focused on the expression of DNA-PKcs in tumor cells [5, 95,96,97]. DNA-PKcs is involved in multiple cellular processes during tumor occurrence and progression, including the cell cycle, proliferation, metastasis, and transcriptional regulation [2]. DNA-PKcs is dysregulated in a variety of tumors, including prostate, breast, and cervical cancers. Genetic or pharmacological approaches have been used to reduce DNA-PK expression in tumor cells, thereby increasing their sensitivity to radiation or chemotherapy agents. The therapeutic effect of DNA-PKcs inhibitors in tumors may be due to their inhibition of the activity of certain transcription factors [4, 7, 11]. Given the oncogenic effect of DNA-PK, inhibitors targeting DNA-PK kinase activity have been developed and tested in clinical trials [4]. Although the in vitro results of the DNA-PK inhibitor experiments were encouraging, the in vivo research results were unsatisfactory, possibly owing to the limited solubility of NU7441 in vivo, which leads to poor absorption in mice. Compared with NU7441, targeting DNA-PK with a clinical-grade dual inhibitor of DNA-PK and TOR kinase (TORK) (CC-115) inhibits the proliferation of CRPC cells and affects DNA-PKcs dependent transcriptional events, including cancer-related pathways such as epithelial-mesenchymal transition and transforming growth factor β (TGFβ) signalling [4].

Though DNA-PKcs is extensively studied in tumors, the specific function of DNA-PKcs in immune cells remains to be elucidated. Owing to the resolution of the tumor microenvironment, cell therapy and immunotherapy play an increasingly important role in the field of precision cancer treatment [98]. DNA-PKcs, a key point in T cell activation, could promote Egr1 transcription factor dependent IL2 transcription [49]. However, combination therapy with regorafenib and NU7441 increased the activation of T cells and co-stimulatory markers as well as cytokines such as IFNγ, TNFα, and IL2 [99]. These studies suggest that the pharmacological inhibition of DNA-PKcs may serve as a method to enhance T cell immunotherapy.

The combination therapy of DNA-PK inhibition and mRNA splicing is a promising strategy. DNA-PKcs could regulate mRNA splicing, and splicing activity is impaired in cells with reduced DNA-PKcs activity. To identify genes that promote DNA-PK inhibitors resistance, researchers conducted CRISPR/Cas9 screening and identified genes related to RNA splicing. Moreover, pladienolide B-mediated splicing inhibition enhances the cytotoxicity of DNA-PK inhibitors in various human cancer cell types. This indicates that the combination therapy of DNA-PK inhibitors and mRNA splicing inhibitors has the potential for tumor treatment in vitro. However, the mechanisms of combination therapy and how RNA splicing related genes promote DNA-PK inhibitors resistance are still unclear.

In conclusion, targeting DNA-PKcs is a promising therapeutic strategy that can produce significant anticancer effects. Further understanding of the functions of DNA-PKcs, particularly transcriptional regulation and RNA metabolism, could provide insights for developing effective therapeutic approaches.

Abbreviations

DNA-PKcs:

DNA-dependent protein kinase catalytic subunit

PIKK:

Phosphatidylinositol 3-kinase-related kinase

NHEJ:

Nonhomologous DNA end-joining

lncRNAs:

Long noncoding RNAs

miRNAs:

Micro RNAs

circRNAs:

Circular RNAs

Lama4:

Laminin alpha 4

Nrf2:

NF-E2-related factor 2

hXOR:

Human xanthine oxidoreductase gene

OSM:

Oncostatin M

TopoIIβ:

DNA topoisomerase IIβ

PARP-1:

Poly [adenosine diphosphate (ADP)–ribose] polymerase–1

Pol II:

RNA polymerase II

DSB:

DNA double-strand break

HIFs:

Hypoxia-inducible factors

TRIM28:

Tripartite motif containing protein 28

HREs:

Hypoxia response elements

CDK9:

Cyclin-dependent kinase 9

CTD:

C-terminal domain

FoxA2:

Forkhead box A2

CRY:

Cryptochrome

PDX-1:

Pancreatic duodenal homeobox-1 protein

EMT:

Epithelial-mesenchymal transition

GSCs:

Glioma stem cells

HSF1:

Heat shock transcription factor 1

PR:

Progesterone receptor

AIRE:

Autoimmune regulatory factor

mTECs:

Myeloid thymic epithelial cells

TLR:

Toll-like receptor

Egr1:

Early growth response protein 1

IRF-3:

IFN regulatory factor 3

FAS:

Fatty acid synthase

WDR6:

WD40 repeat-containing protein 6

USF-1:

Upstream stimulatory factor 1

Zic2:

Zinc finger protein of cerebellum 2

RHA:

RNA helicase A

NF-κB:

Nuclear factor-kappa B

NEMO:

NF-κB essential modulator

EGFR:

Epidermal growth factor receptor

MYT1L:

Myelin transcription factor 1-like

LEF1:

Lymphoid enhancer binding factor 1

CRPC:

Castration-resistant prostate cancer

DDR:

DNA damage repair

YB-1:

Y-box binding protein

BCLAF1:

Bcl-2-associated transcription factor 1

KLF8:

Krüppel-like factor 8

NR4A:

Nuclear receptor subfamily 4 group A

SRF:

Serum response factor

MTX:

Methotrexate

AS:

Alternative splicing

RBMX:

RNA binding motif protein X-linked

HEAT:

Huntingtin, elongation factor 3, PP2A, and TOR1

TCR:

T cell receptor; transcription-coupled repair

References

  1. Dylgjeri E, Knudsen KE. DNA-PKcs: a targetable protumorigenic protein kinase. Cancer Res. 2022;82(4):523–33.

    Article  CAS  PubMed  Google Scholar 

  2. Goodwin JF, Knudsen KE. Beyond DNA repair: DNA-PK function in cancer. Cancer Discov. 2014;4(10):1126–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hsu FM, Zhang S, Chen BP. Role of DNA-dependent protein kinase catalytic subunit in cancer development and treatment. Transl Cancer Res. 2012;1(1):22–34.

    PubMed  Google Scholar 

  4. Dylgjeri E, McNair C, Goodwin JF, Raymon HK, McCue PA, Shafi AA, et al. Pleiotropic Impact of DNA-PK in cancer and implications for therapeutic strategies. Clin Cancer Res. 2019;25(18):5623–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ciszewski WM, Tavecchio M, Dastych J, Curtin NJ. DNA-PK inhibition by NU7441 sensitizes breast cancer cells to ionizing radiation and doxorubicin. Breast Cancer Res Treat. 2014;143(1):47–55.

    Article  CAS  PubMed  Google Scholar 

  6. Jackson SP, MacDonald JJ, Lees-Miller S, Tjian R. GC box binding induces phosphorylation of Sp1 by a DNA-dependent protein kinase. Cell. 1990;63(1):155–65.

    Article  CAS  PubMed  Google Scholar 

  7. Yang Y, Lu H, Chen C, Lyu Y, Cole RN, Semenza GL. HIF-1 interacts with TRIM28 and DNA-PK to release paused RNA polymerase II and activate target gene transcription in response to hypoxia. Nat Commun. 2022;13(1):316.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chen Y, Schnetz MP, Irarrazabal CE, Shen RF, Williams CK, Burg MB, et al. Proteomic identification of proteins associated with the osmoregulatory transcription factor TonEBP/OREBP: functional effects of Hsp90 and PARP-1. Am J Physiol Renal Physiol. 2007;292(3):F981–92.

    Article  CAS  PubMed  Google Scholar 

  9. Liu S, Shao Y, Wang Q, Zhai Y, Li X. Genotoxic stress causes the accumulation of DNA-dependent protein kinase catalytic subunit phosphorylated at serine 2056 at nuclear speckles and alters pre-mRNA alternative splicing. FEBS Open Bio. 2019;9(2):304–14.

    Article  CAS  PubMed  Google Scholar 

  10. Woodard RL, Anderson MG, Dynan WS. Nuclear extracts lacking DNA-dependent protein kinase are deficient in multiple round transcription. J Biol Chem. 1999;274(1):478–85.

    Article  CAS  PubMed  Google Scholar 

  11. Goodwin JF, Kothari V, Drake JM, Zhao S, Dylgjeri E, Dean JL, et al. DNA-PKcs-mediated transcriptional regulation drives prostate cancer progression and metastasis. Cancer Cell. 2015;28(1):97–113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li K, Zhang S, Gu Y, Wang J, Yang Y, Mao W. Transcriptomic data of BT549 triple negative breast cancer cells treated with 20 µM NU7441, a DNA-dependent kinase inhibitor. Data Brief. 2024;53: 110183.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bryntesson F, Regan JC, Jeggo PA, Taccioli GE, Hubank M. Analysis of gene transcription in cells lacking DNA-PK activity. Radiat Res. 2001;156(2):167–76.

    Article  CAS  PubMed  Google Scholar 

  14. Xu X, Zhang Y, Li W, Miao H, Zhang H, Zhou Y, et al. Wogonin reverses multi-drug resistance of human myelogenous leukemia K562/A02 cells via downregulation of MRP1 expression by inhibiting Nrf2/ARE signaling pathway. Biochem Pharmacol. 2014;92(2):220–34.

    Article  CAS  PubMed  Google Scholar 

  15. Lin J, Xu P, LaVallee P, Hoidal JR. Identification of proteins binding to E-Box/Ku86 sites and function of the tumor suppressor SAFB1 in transcriptional regulation of the human xanthine oxidoreductase gene. J Biol Chem. 2008;283(44):29681–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wang B, Li D, Yao Y, Heyns M, Kovalchuk A, Ilnytskyy Y, et al. The crucial role of DNA-dependent protein kinase and myelin transcription factor 1-like protein in the miR-141 tumor suppressor network. Cell Cycl. 2019;18(21):2876–92.

    Article  CAS  Google Scholar 

  17. Kothari V, Goodwin JF, Zhao SG, Drake JM, Yin Y, Chang SL, et al. DNA-dependent protein kinase drives prostate cancer progression through transcriptional regulation of the Wnt signaling pathway. Clin Cancer Res. 2019;25(18):5608–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ju BG, Lunyak VV, Perissi V, Garcia-Bassets I, Rose DW, Glass CK, et al. A topoisomerase IIbeta-mediated dsDNA break required for regulated transcription. Science. 2006;312(5781):1798–802.

    Article  CAS  PubMed  Google Scholar 

  19. Ju BG, Rosenfeld MG. A breaking strategy for topoisomerase IIbeta/PARP-1-dependent regulated transcription. Cell Cycl. 2006;5(22):2557–60.

    Article  CAS  Google Scholar 

  20. Bunch H, Lawney BP, Lin YF, Asaithamby A, Murshid A, Wang YE, et al. Transcriptional elongation requires DNA break-induced signalling. Nat Commun. 2015;6:10191.

    Article  CAS  PubMed  Google Scholar 

  21. Maldonado E, Shiekhattar R, Sheldon M, Cho H, Drapkin R, Rickert P, et al. A human RNA polymerase II complex associated with SRB and DNA-repair proteins. Nature. 1996;381(6577):86–9.

    Article  CAS  PubMed  Google Scholar 

  22. Caron P, Pankotai T, Wiegant WW, Tollenaere MAX, Furst A, Bonhomme C, et al. WWP2 ubiquitylates RNA polymerase II for DNA-PK-dependent transcription arrest and repair at DNA breaks. Genes Dev. 2019;33(11–12):684–704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Pankotai T, Bonhomme C, Chen D, Soutoglou E. DNAPKcs-dependent arrest of RNA polymerase II transcription in the presence of DNA breaks. Nat Struct Mol Biol. 2012;19(3):276–82.

    Article  CAS  PubMed  Google Scholar 

  24. Bossaert M, Moreno A, Peixoto A, Pillaire MJ, Chanut P, Frit P, et al. Identification of the main barriers to Ku accumulation in chromatin. bioRxiv. 2024;3:2033.

    Google Scholar 

  25. Nueda A, Hudson F, Mivechi NF, Dynan WS. DNA-dependent protein kinase protects against heat-induced apoptosis. J Biol Chem. 1999;274(21):14988–96.

    Article  CAS  PubMed  Google Scholar 

  26. Treviño LS, Bolt MJ, Grimm SL, Edwards DP, Mancini MA, Weigel NL. Differential regulation of progesterone receptor-mediated transcription by CDK2 and DNA-PK. Mol Endocrinol. 2016;30(2):158–72.

    Article  PubMed  Google Scholar 

  27. Schild-Poulter C, Shih A, Tantin D, Yarymowich NC, Soubeyrand S, Sharp PA, et al. DNA-PK phosphorylation sites on Oct-1 promote cell survival following DNA damage. Oncogene. 2007;26(27):3980–8.

    Article  CAS  PubMed  Google Scholar 

  28. Schild-Poulter C, Shih A, Yarymowich NC, Haché RJ. Down-regulation of histone H2B by DNA-dependent protein kinase in response to DNA damage through modulation of octamer transcription factor 1. Cancer Res. 2003;63(21):7197–205.

    CAS  PubMed  Google Scholar 

  29. Nock A, Ascano JM, Jones T, Barrero MJ, Sugiyama N, Tomita M, et al. Identification of DNA-dependent protein kinase as a cofactor for the forkhead transcription factor FoxA2. J Biol Chem. 2009;284(30):19915–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. de Ruyck J, Brysbaert G, Villeret V, Aumercier M, Lensink MF. Computational characterization of the binding mode between oncoprotein Ets-1 and DNA-repair enzymes. Proteins. 2018;86(10):1055–63.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Choul-Li S, Legrand AJ, Bidon B, Vicogne D, Villeret V, Aumercier M. Ets-1 interacts through a similar binding interface with Ku70 and poly (ADP-Ribose) polymerase-1. Biosci Biotechnol Biochem. 2018;82(10):1753–9.

    Article  CAS  PubMed  Google Scholar 

  32. Brenner JC, Ateeq B, Li Y, Yocum AK, Cao Q, Asangani IA, et al. Mechanistic rationale for inhibition of poly(ADP-ribose) polymerase in ETS gene fusion-positive prostate cancer. Cancer Cell. 2011;19(5):664–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Piggin CL, Roden DL, Law AMK, Molloy MP, Krisp C, Swarbrick A, et al. ELF5 modulates the estrogen receptor cistrome in breast cancer. PLoS Genet. 2020;16(1): e1008531.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ishiguro A, Ideta M, Mikoshiba K, Chen DJ, Aruga J. ZIC2-dependent transcriptional regulation is mediated by DNA-dependent protein kinase, poly(ADP-ribose) polymerase, and RNA helicase A. J Biol Chem. 2007;282(13):9983–95.

    Article  CAS  PubMed  Google Scholar 

  35. Ishiguro A, Aruga J. Functional role of Zic2 phosphorylation in transcriptional regulation. FEBS Lett. 2008;582(2):154–8.

    Article  CAS  PubMed  Google Scholar 

  36. Wong RH, Chang I, Hudak CS, Hyun S, Kwan HY, Sul HS. A role of DNA-PK for the metabolic gene regulation in response to insulin. Cell. 2009;136(6):1056–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gao P, Yoo SH, Lee KJ, Rosensweig C, Takahashi JS, Chen BP, et al. Phosphorylation of the cryptochrome 1 C-terminal tail regulates circadian period length. J Biol Chem. 2013;288(49):35277–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu SH, Ma JT, Yueh AY, Lees-Miller SP, Anderson CW, Ng SY. The carboxyl-terminal transactivation domain of human serum response factor contains DNA-activated protein kinase phosphorylation sites. J Biol Chem. 1993;268(28):21147–54.

    Article  CAS  PubMed  Google Scholar 

  39. Bannister AJ, Gottlieb TM, Kouzarides T, Jackson SP. c-Jun is phosphorylated by the DNA-dependent protein kinase in vitro; definition of the minimal kinase recognition motif. Nucl Acid Res. 1993;21(5):1289–95.

    Article  CAS  Google Scholar 

  40. Liu Z, Merkurjev D, Yang F, Li W, Oh S, Friedman MJ, et al. Enhancer activation requires trans-recruitment of a mega transcription factor complex. Cell. 2014;159(2):358–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sutcu HH, Montagne B, Ricchetti M. DNA-PKcs regulates myogenesis in an Akt-dependent manner independent of induced DNA damage. Cell Death Differ. 2023;30(8):1900–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zou R, Shi W, Chang X, Zhang M, Tan S, Li R, et al. The DNA-dependent protein kinase catalytic subunit exacerbates endotoxemia-induced myocardial microvascular injury by disrupting the MOTS-c/JNK pathway and inducing profilin-mediated lamellipodia degradation. Theranostics. 2024;14(4):1561–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lebrun P, Montminy MR, Van Obberghen E. Regulation of the pancreatic duodenal homeobox-1 protein by DNA-dependent protein kinase. J Biol Chem. 2005;280(46):38203–10.

    Article  CAS  PubMed  Google Scholar 

  44. Bouquet F, Ousset M, Biard D, Fallone F, Dauvillier S, Frit P, et al. A DNA-dependent stress response involving DNA-PK occurs in hypoxic cells and contributes to cellular adaptation to hypoxia. J Cell Sci. 2011;124(Pt 11):1943–51.

    Article  CAS  PubMed  Google Scholar 

  45. Pyun BJ, Seo HR, Lee HJ, Jin YB, Kim EJ, Kim NH, et al. Mutual regulation between DNA-PKcs and Snail1 leads to increased genomic instability and aggressive tumor characteristics. Cell Death Dis. 2013;4(2): e517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. An J, Yang DY, Xu QZ, Zhang SM, Huo YY, Shang ZF, et al. DNA-dependent protein kinase catalytic subunit modulates the stability of c-Myc oncoprotein. Mol Cancer. 2008;7:32.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Fang X, Huang Z, Zhai K, Huang Q, Tao W, Kim L, et al. Inhibiting DNA-PK induces glioma stem cell differentiation and sensitizes glioblastoma to radiation in mice. Sci Transl Med. 2021. https://doi.org/10.1126/scitranslmed.abc7275.

    Article  PubMed  Google Scholar 

  48. Ghonim MA, Pyakurel K, Ju J, Rodriguez PC, Lammi MR, Davis C, et al. DNA-dependent protein kinase inhibition blocks asthma in mice and modulates human endothelial and CD4(+) T-cell function without causing severe combined immunodeficiency. J Allerg Clin Immunol. 2015;135(2):425–40.

    Article  CAS  Google Scholar 

  49. Waldrip ZJ, Burdine L, Harrison DK, Azevedo-Pouly AC, Storey AJ, Moffett OG, et al. DNA-PKcs kinase activity stabilizes the transcription factor Egr1 in activated immune cells. J Biol Chem. 2021;297(4): 101209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ferguson BJ, Mansur DS, Peters NE, Ren H, Smith GL. DNA-PK is a DNA sensor for IRF-3-dependent innate immunity. Elife. 2012;1: e00047.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Karpova AY, Trost M, Murray JM, Cantley LC, Howley PM. Interferon regulatory factor-3 is an in vivo target of DNA-PK. Proc Natl Acad Sci USA. 2002;99(5):2818–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Liiv I, Rebane A, Org T, Saare M, Maslovskaja J, Kisand K, et al. DNA-PK contributes to the phosphorylation of AIRE: importance in transcriptional activity. Biochim Biophys Acta. 2008;1783(1):74–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Abramson J, Giraud M, Benoist C, Mathis D. Aire’s partners in the molecular control of immunological tolerance. Cell. 2010;140(1):123–35.

    Article  CAS  PubMed  Google Scholar 

  54. Mathieu AL, Verronese E, Rice GI, Fouyssac F, Bertrand Y, Picard C, et al. PRKDC mutations associated with immunodeficiency, granuloma, and autoimmune regulator-dependent autoimmunity. J Allerg Clin Immunol. 2015;135(6):1578-88.e5.

    Article  CAS  Google Scholar 

  55. Wu J, Zhu W, Fu H, Zhang Y, Sun J, Yang W, et al. DNA-PKcs interacts with Aire and regulates the expression of toll-like receptors in RAW264.7 cells. Scand J Immunol. 2012;75(5):479–88.

    Article  CAS  PubMed  Google Scholar 

  56. Hasegawa Y, Asada S. DNA-dependent protein kinase catalytic subunit binds to the transactivation domain 1 of NF-κB p65. Biochem Biophys Rep. 2023;35: 101538.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Medunjanin S, Putzier M, Nöthen T, Weinert S, Kähne T, Luani B, et al. DNA-PK: gatekeeper for IKKγ/NEMO nucleocytoplasmic shuttling in genotoxic stress-induced NF-kappaB activation. Cell Mol Life Sci. 2020;77(20):4133–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Sabatel H, Pirlot C, Piette J, Habraken Y. Importance of PIKKs in NF-κB activation by genotoxic stress. Biochem Pharmacol. 2011;82(10):1371–83.

    Article  CAS  PubMed  Google Scholar 

  59. Nöthen T, Sarabi MA, Weinert S, Zuschratter W, Morgenroth R, Mertens PR, et al. DNA-dependent protein kinase mediates YB-1 (Y-Box binding protein)-induced double strand break repair. Arterioscler Thromb Vasc Biol. 2023;43(2):300–11.

    Article  PubMed  Google Scholar 

  60. Lee YY, Yu YB, Gunawardena HP, Xie L, Chen X. BCLAF1 is a radiation-induced H2AX-interacting partner involved in γH2AX-mediated regulation of apoptosis and DNA repair. Cell Death Dis. 2012;3(7): e359.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lu H, Hu L, Li T, Lahiri S, Shen C, Wason MS, et al. A novel role of Krüppel-like factor 8 in DNA repair in breast cancer cells. J Biol Chem. 2012;287(52):43720–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Malewicz M, Kadkhodaei B, Kee N, Volakakis N, Hellman U, Viktorsson K, et al. Essential role for DNA-PK-mediated phosphorylation of NR4A nuclear orphan receptors in DNA double-strand break repair. Genes Dev. 2011;25(19):2031–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Haemmig S, Yang D, Sun X, Das D, Ghaffari S, Molinaro R, et al. Long noncoding RNA SNHG12 integrates a DNA-PK-mediated DNA damage response and vascular senescence. Sci Transl Med. 2020. https://doi.org/10.1126/scitranslmed.aaw1868.

    Article  PubMed  Google Scholar 

  64. Guo Z, Wang YH, Xu H, Yuan CS, Zhou HH, Huang WH, et al. LncRNA linc00312 suppresses radiotherapy resistance by targeting DNA-PKcs and impairing DNA damage repair in nasopharyngeal carcinoma. Cell Death Dis. 2021;12(1):69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wang X, Liu H, Shi L, Yu X, Gu Y, Sun X. LINP1 facilitates DNA damage repair through nonhomologous end joining (NHEJ) pathway and subsequently decreases the sensitivity of cervical cancer cells to ionizing radiation. Cell Cycl. 2018;17(4):439–47.

    Article  CAS  Google Scholar 

  66. Taiana E, Bandini C, Favasuli VK, Ronchetti D, Silvestris I, Puccio N, et al. Activation of long noncoding RNA NEAT1 leads to survival advantage of multiple myeloma cells by supporting a positive regulatory loop with DNA repair proteins. Haematologica. 2023;108(1):219–33.

    Article  CAS  PubMed  Google Scholar 

  67. Morchikh M, Cribier A, Raffel R, Amraoui S, Cau J, Severac D, et al. HEXIM1 and NEAT1 long noncoding RNA form a multi-subunit complex that regulates DNA-mediated innate immune response. Mol Cell. 2017;67(3):387-99.e5.

    Article  CAS  PubMed  Google Scholar 

  68. Zhang Y, He Q, Hu Z, Feng Y, Fan L, Tang Z, et al. Long noncoding RNA LINP1 regulates repair of DNA double-strand breaks in triple-negative breast cancer. Nat Struct Mol Biol. 2016;23(6):522–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhang H, Ma RR, Zhang G, Dong Y, Duan M, Sun Y, et al. Long noncoding RNA lnc-LEMGC combines with DNA-PKcs to suppress gastric cancer metastasis. Cancer Lett. 2022;524:82–90.

    Article  CAS  PubMed  Google Scholar 

  70. Spurlock CF 3rd, Tossberg JT, Matlock BK, Olsen NJ, Aune TM. Methotrexate inhibits NF-κB activity via long intergenic (noncoding) RNA-p21 induction. Arthritis Rheumatol. 2014;66(11):2947–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lyu F, Huang S, Yan Z, He Q, Liu C, Cheng L, et al. CircUGGT2 facilitates progression and cisplatin resistance of bladder cancer through nonhomologous end-joining pathway. Cell Signal. 2024;119: 111164.

    Article  PubMed  Google Scholar 

  72. Tan L, Huang Z, Chen Z, Chen S, Ye Y, Chen T, et al. CircRNA_001895 promotes sunitinib resistance of renal cell carcinoma through regulation of apoptosis and DNA damage repair. J Chemother. 2023;35(1):11–8.

    Article  CAS  PubMed  Google Scholar 

  73. Li N, Ma Y, Ma L, Guan Y, Ma L, Yang D. MicroRNA-488-3p sensitizes malignant melanoma cells to cisplatin by targeting PRKDC. Cell Biol Int. 2017;41(6):622–9.

    Article  CAS  PubMed  Google Scholar 

  74. Sudhanva MS, Hariharasudhan G, Jun S, Seo G, Kamalakannan R, Kim HH, et al. MicroRNA-145 impairs classical nonhomologous end-joining in response to ionizing radiation-induced DNA double-strand breaks via targeting DNA-PKcs. Cells. 2022. https://doi.org/10.3390/cells11091509.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Li Y, Han W, Ni TT, Lu L, Huang M, Zhang Y, et al. Knockdown of microRNA-1323 restores sensitivity to radiation by suppression of PRKDC activity in radiation-resistant lung cancer cells. Oncol Rep. 2015;33(6):2821–8.

    Article  CAS  PubMed  Google Scholar 

  76. Lee KM, Choi EJ, Kim IA. microRNA-7 Increases radiosensitivity of human cancer cells with activated EGFR-associated signaling. Radiother Oncol. 2011;101(1):171–6.

    Article  CAS  PubMed  Google Scholar 

  77. Chen S, Wang H, Ng WL, Curran WJ, Wang Y. Radiosensitizing effects of ectopic miR-101 on non-small cell lung cancer cells depend on the endogenous miR-101 level. Int J Radiat Oncol Biol Phys. 2011;81(5):1524–9.

    Article  CAS  PubMed  Google Scholar 

  78. Gu H, Li J, Zhang R. Melatonin upregulates DNA-PKcs to suppress apoptosis of human umbilical vein endothelial cells via inhibiting miR-101 under H(2)O(2)-induced oxidative stress. Mol Cell Biochem. 2021;476(2):1283–92.

    Article  CAS  PubMed  Google Scholar 

  79. Li W, Guo F, Wang P, Hong S, Zhang C. miR-221/222 Confers radioresistance in glioblastoma cells through activating Akt independent of PTEN status. Curr Mol Med. 2014;14(1):185–95.

    Article  CAS  PubMed  Google Scholar 

  80. Bergstrand S, O’Brien EM, Coucoravas C, Hrossova D, Peirasmaki D, Schmidli S, et al. Small Cajal body-associated RNA 2 (scaRNA2) regulates DNA repair pathway choice by inhibiting DNA-PK. Nat Commun. 2022;13(1):1015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Shao Z, Flynn RA, Crowe JL, Zhu Y, Liang J, Jiang W, et al. DNA-PKcs has KU-dependent function in rRNA processing and haematopoiesis. Nature. 2020;579(7798):291–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Li P, Gai X, Li Q, Yang Q, Yu X. DNA-PK participates in pre-rRNA biogenesis independent of DNA double-strand break repair. Nucl Acid Res. 2024;52(11):6360–75.

    Article  CAS  Google Scholar 

  83. Adamson B, Brittain N, Walker L, Duncan R, Luzzi S, Rescigno P, et al. The catalytic subunit of DNA-PK regulates transcription and splicing of AR in advanced prostate cancer. J Clin Invest. 2023. https://doi.org/10.1172/JCI169200.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Song Z, Xie Y, Guo Z, Han Y, Guan H, Liu X, et al. Genome-wide identification of DNA-PKcs-associated RNAs by RIP-Seq. Signal Transduct Target Ther. 2019;4:22.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Camfield S, Chakraborty S, Dwivedi SKD, Pramanik PK, Mukherjee P, Bhattacharya R. Secrets of DNA-PKcs beyond DNA repair. NPJ Precis Oncol. 2024;8(1):154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Calkins AS, Iglehart JD, Lazaro JB. DNA damage-induced inhibition of rRNA synthesis by DNA-PK and PARP-1. Nucl Acid Res. 2013;41(15):7378–86.

    Article  CAS  Google Scholar 

  87. Cerqueira AV, Lemos B. Ribosomal DNA and the nucleolus as keystones of nuclear architecture, organization, and function. Trend Genet. 2019;35(10):710–23.

    Article  CAS  Google Scholar 

  88. Hein N, Hannan KM, George AJ, Sanij E, Hannan RD. The nucleolus: an emerging target for cancer therapy. Trends Mol Med. 2013;19(11):643–54.

    Article  CAS  PubMed  Google Scholar 

  89. Milano L, Gautam A, Caldecott KW. DNA damage and transcription stress. Mol Cell. 2024;84(1):70–9.

    Article  CAS  PubMed  Google Scholar 

  90. Hammarsten O, Chu G. DNA-dependent protein kinase: DNA binding and activation in the absence of Ku. Proc Natl Acad Sci USA. 1998;95(2):525–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. West RB, Yaneva M, Lieber MR. Productive and nonproductive complexes of Ku and DNA-dependent protein kinase at DNA termini. Mol Cell Biol. 1998;18(10):5908–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Yin X, Liu M, Tian Y, Wang J, Xu Y. Cryo-EM structure of human DNA-PK holoenzyme. Cell Res. 2017;27(11):1341–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Chen X, Xu X, Chen Y, Cheung JC, Wang H, Jiang J, et al. Structure of an activated DNA-PK and its implications for NHEJ. Mol Cell. 2021. https://doi.org/10.1016/j.molcel.2020.12.015.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Schild-Poulter C, Pope L, Giffin W, Kochan JC, Ngsee JK, Traykova-Andonova M, et al. The binding of Ku antigen to homeodomain proteins promotes their phosphorylation by DNA-dependent protein kinase. J Biol Chem. 2001;276(20):16848–56.

    Article  CAS  PubMed  Google Scholar 

  95. Olsen BB, Issinger OG, Guerra B. Regulation of DNA-dependent protein kinase by protein kinase CK2 in human glioblastoma cells. Oncogene. 2010;29(45):6016–26.

    Article  CAS  PubMed  Google Scholar 

  96. Dibitetto D, Marshall S, Sanchi A, Liptay M, Badar J, Lopes M, et al. DNA-PKcs promotes fork reversal and chemoresistance. Mol Cell. 2022;82(20):3932-42.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Mishra SV, Banerjee A, Sarkar D, Thangarathnam V, Bagal B, Hasan SK, et al. DNA-PKcs-mediated transcriptional regulation of TOP2B drives chemoresistance in acute myeloid leukemia. J Cell Sci. 2024. https://doi.org/10.1242/jcs.261931.

    Article  PubMed  Google Scholar 

  98. Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G, Zitvogel L. Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 2016;27(8):1482–92.

    Article  CAS  PubMed  Google Scholar 

  99. Tsai AK, Khan AY, Worgo CE, Wang LL, Liang Y, Davila E. A multikinase and DNA-PK inhibitor combination immunomodulates melanomas, suppresses tumor progression, and enhances immunotherapies. Cancer Immunol Res. 2017;5(9):790–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This study was supported by the National Natural Science Foundation of China (grant no. 82273130 to T.M.) and the Beijing Municipal Public Welfare Development and Reform Pilot Project for Medical Research Institutes (PWD&RPP-MRI; project no. JYY2023-14).

Author information

Authors and Affiliations

Authors

Contributions

Concept and design: T.M. and J.B.H. Data analysis and interpretation: J.B.H. Manuscript writing: all authors. Final approval of the manuscript: all authors.

Corresponding authors

Correspondence to Ping-Kun Zhou or Teng Ma.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hou, J., Lu, M., Guo, J. et al. DNA-PKcs, a player winding and dancing with RNA metabolism and diseases. Cell Mol Biol Lett 30, 25 (2025). https://doi.org/10.1186/s11658-025-00703-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s11658-025-00703-z

Keywords