Open Access

HELQ as a DNA helicase: Its novel role in normal cell function and tumorigenesis (Review)

  • Authors:
    • Nan Tang
    • Weilun Wen
    • Zhihe Liu
    • Xifeng Xiong
    • Yanhua Wu
  • View Affiliations

  • Published online on: November 2, 2023     https://doi.org/10.3892/or.2023.8657
  • Article Number: 220
  • Copyright: © Tang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Helicase POLQ‑like (HELQ or Hel308), is a highly conserved, 3'‑5' superfamily II DNA helicase that contributes to diverse DNA processes, including DNA repair, unwinding, and strand annealing. HELQ deficiency leads to subfertility, due to its critical role in germ cell stability. In addition, the abnormal expression of HELQ has been observed in multiple tumors and a number of molecular pathways, including the nucleotide excision repair, checkpoint kinase 1‑DNA repair protein RAD51 homolog 1 and ATM/ATR pathways, have been shown to be involved in HELQ. In the present review, the structure and characteristics of HELQ, as well as its major functions in DNA processing, were described. Molecular mechanisms involving HELQ in the context of tumorigenesis were also described. It was deduced that HELQ biology warrants investigation, and that its critical roles in the regulation of various DNA processes and participation in tumorigenesis are clinically relevant.

Introduction

The Helicase POLQ-like (HELQ) gene, also referred to as Hel308/holliday junction migration protein, maps to human chromosome 4q21 and was first cloned by its homology to Drosophila melanogaster (D. melanogaster) mutagen-sensitive 308 (Mus308) (1). In humans, HELQ is typical superfamily-2 (SF2) ATPase-dependent 3′ to 5′ helicase as a functions to unwind DNA (2,3). As HELQ has ATPase and helicase functions, it has been classified as a member of the Ski2-like subfamily of SF2 helicases (4), which is a small subfamily (5,6) involved in RNA processing, splicing and degradation pathways (7). Similar to other SF2 helicases, HELQ is a regulator of genome stability (2), DNA recombination (2), DNA binding (8), DNA unwinding (9), DNA replication (10), DNA strand annealing (9) and DNA repair (11,12).

HELQ is widely expressed in normal human tissues, particularly of the reproductive system, and plays an important role in maintaining germ cells by promoting DNA repair protein RecA-like ATP-dependent recombinase 51 (RAD51) homolog 1 (RAD51) paralog-dependent repair (11). Furthermore, HELQ is abnormally expressed in multiple tumors, and contributes to cell proliferation, metastasis and chemotherapy resistance through various molecular pathways, such as nucleotide excision repair (NER), as well as the checkpoint kinase 1 (CHK1)-RAD51 and ataxia telangiectasia mutated protein/ataxia telangiectasia and Rad3-related protein (ATM/ATR) signaling pathways (12,13). HELQ can also contribute to germ cell maintenance and tumorigenesis in mammals by regulating its target genes, which include CHK1, fanconi anemia group D2 (FANCD2), RAD51 and replication protein A (RPA), among others (3,4,9,14). Of note, the low expression or loss of HELQ can have a significant impact on clinical features and prognosis in patients with cancer.

In the present review, the structural features, function, target genes, roles in reproduction, molecular mechanisms associated with tumorigenesis, and potential applications of HELQ were summarized. It was concluded that HELQ is an attractive future clinical target in conditions involving germ cell maintenance and in molecular targeted cancer therapies.

Structural features of HELQ

SF2 helicase family

The DNA helicases are classified into five different superfamilies (SF1-SF5) (15). SF1 and SF2 helicases encompass a large group of DNA and RNA helicases found in eubacteria, archaea, viruses and eukaryotes. They possess an ATP-dependent translocation module consisting of two RecA-fold domains responsible for nucleic acid and ATP binding (15,16). HELQ was first described by Marini in 2002, which belongs to the SF2 protein family (1). SF2 family proteins have a pair of RecA-like domains that provide motion associated with helicase activity (6,1719) and play key roles in chromatin rearrangement, DNA repair and transcription (2022) and RNA metabolism (17,23). SF2 family proteins are divided into Ski2-like, RecQ-like, RecG-like, RecA-like ATP-dependent recombinase 3 (Rad3)/XPD, type I restriction enzymes, DEAD-box and NS3/NPH-II subfamilies, Swi/Snf, DEAH/RNA helicase A, RIG-I-like, based on sequence homology (6,24,25). In archaea, in all Ski2-like helicases including Mtr4, Ski2, and Bad Response to Refrigeration 2 homolog, the molecular ‘core’ of them is a ring-like assembly of four domains comprising a ratchet domain, a winged helix domain and two RecA domains (25).

Structural and functional characteristics of HELQ

HELQ maps to chromosome 4q21 (1) and generates a full-length mRNA (NM_133636.5) comprising 3,543 base pairs and encoding a protein of 1,101 amino acids (aa) (1). A total of 7 transcribed splice isoforms of HELQ have been described, encoding six HELQ protein variants (26). HELQ is a conserved protein that possesses three main protein domains: The DEAD/DEAH box helicase domain, a helix-turn-helix (HTH_61) domain and a helicase C-terminal domain (Fig. 1). The domain of DEAD/DEAH box helicase contains a DEAH box and an ATP-binding region (putative ATP binding site), and is involved in unwinding nucleic acids. In addition, human HELQ has a disordered domain (212–261 aa), containing two compositionally biased regions, comprising basic and acidic (214–228 aa) and polar (229–253 aa) residues. A structural diagram of the human HELQ protein is presented in Fig. 1.

The DEAD/DEAH box helicase class of proteins, which share eight conserved motifs (I, Ia, Ib, II, III, IV, V and VI) (27). ATP is bound to motifs I and II, while RNA is bound to motifs Ia and Ib. The DEAD/DEAH box is named after the Walker B motif (motif III) consensus sequence (28) and functions in ATP hydrolysis. Motifs IV and V perform similar functions to those of motifs Ia and Ib, respectively, in RNA binding (27). DEAD/DEAH box helicases are found in various prokaryotes and eukaryotes (28), and are involved in several aspects of RNA metabolism, such as pre-mRNA splicing, RNA decay, nuclear transcription, editing, nucleocytoplasmic transport, ribosome biogenesis, translation and organellar gene expression (2931).

The helicase C-terminal domain (HelicC) is present in various SF1 and SF2 helicases and helicase-related proteins. The HelicC domain does not fold autonomously, but rather as a component of the helicase, which participates in the ATP-dependent unwinding of DNA or RNA.

The α-helical protein domain family includes winged helix-turn-helix (HTH) domains that have characteristic folds, which function as sequence-specific DNA-binding domains (32). The HTH plays an important role in DNA binding and protein interactions (33,34) and consists of two α-helices (α-helix 22 and 23) and a β-sheet turn, where a double-stranded DNA (dsDNA) major groove can be recognized by α-helix 23 (14,2).

HELQ expression pattern

HELQ is highly conserved across a wide range of species from archaea through to mammals (https://www.ncbi.nlm.nih.gov/homologene/?term=HELQ). For example, Homo sapiens HELQ shares a >97, 78 and 60% DNA similarity with the homologous Macaca mulatta, Mus musculus, and Danio rerio genes, respectively (Table I). It is a conserved gene in eukaryotes that HELQ (HomoloGene ID: 14667) has important molecular functions (i.e., ATP binding, DNA binding, single-stranded 3′-5′DNA helicase activity) and cellular component classifications (i.e., nucleus, site of DNA damage), and acts in biological processes (i.e., DNA duplex unwinding, rRNA processing) (1,9,3537) (https://www.alliancegenome.org/gene/HGNC:18536) (Table II). Since HELQ is expressed in a wide variety of species, it was likely present in a common ancestor of vertebrates.

Table I.

Conserved gene HELQ homology in Eukaryota (HomoloGene: 14667) [https://www.ncbi.nlm.nih.gov/homologene/?term=HELQ].

Table I.

Conserved gene HELQ homology in Eukaryota (HomoloGene: 14667) [https://www.ncbi.nlm.nih.gov/homologene/?term=HELQ].

HomoloGene: 14667. Gene conserved in Eukaryota

SpeciesSymbolGenetic locationProtein Acc.Protein lengthIdentity (%)* proteinDNA
H. sapiensHELQChr4 q21.23NP_598375.21101 aa
P. troglodytesHELQChr4XP_003310356.11101 aa99.199.4
M. mulattaHELQChr5 XP-_001104832.11101 aa97.097.7
C. lupusHELQChr32XP_544959.21072 aa85.988.4
B. taurusHELQChr6XP_002688448.31094 aa83.786.3
M. musculusHelqChr5 E4NP_001074576.11069 aa78.679.3
R. norvegicusHelqChr14 p22NP_001014156.21065 aa79.179.0
G. gallusHELQChr4XP_420565.11048 aa68.869.7
X. tropicalishelqChr1XP_002939628.21000 aa66.066.8
D. reriohelqChr21XP_691411.31010 aa60.961.6
D. melanogastermus301Chr3-22cMNP_648178.11051 aa45.850.5
A. gambiae AgaP-AGAP012297Chr3LXP_551895.3  914 aa44.448.3
C. eleganshelq-1ChrIIINP_001022911.1  923 aa41.249.9

[i] HELQ, helicase POLQ-like.

Table II.

Function-Gene Ontology Annotations of HELQ (https://www.alliancegenome.org/gene/HGNC:18536).

Table II.

Function-Gene Ontology Annotations of HELQ (https://www.alliancegenome.org/gene/HGNC:18536).

CategoryClassification termGene Ontology IDaReference
Molecular functionATP bindingGO:0005524GO_REF:0000043
ATP hydrolysis activityGO:0016887GO_REF:0000116
DNA bindingGO:0003677GO_REF:0000043
Single-stranded 3′-5′ DNA helicase activityGO:1990518(1,9,35)
Cellular componentNucleusGO:0005634(9,36)
Site of DNA damageGO:0090734(9)
DNA double-strand break processing involved in repair via single-strand annealingGO:0010792(9)
DNA duplex unwindingGO:0032508GO_REF:0000108
Double-strand break repair via alternative non-homologous end joiningGO:0097681(9)
Biological processDouble-strand break repair via homologous recombinationGO:0000724(37)
Double-strand break repair via synthesis-dependent strand annealingGO:0045003(9)
Positive regulation of double-strand break repair via Homologous recombinationGO:1905168GO_REF:0000024
rRNA processingGO:0006364(36)

a http://amigo.geneontology.org/amigo.

Physiological functions of HELQ

DNA helicases are ubiquitous in living organisms, where they facilitate processes involved in DNA metabolism through unwinding the DNA double helix (3). It is essential for the maintenance of genome stability for DNA helicases to function in the replication and repair of DNA (38). HELQ is a single-stranded 3′-5′ DNA helicase with critical roles in DNA repair, binding, unwinding, replication and strand annealing (2,8,9).

DNA repair

DNA damage is a common event that may have either endogenous or exogenous causes and can lead to mutations, cell/organ death and cancer (39,40). It is crucial to repair damaged DNA after it has been damaged, as this allows damaged DNA to regain its original structure and function normally (34,41). Various pathways recognize and repair different types of DNA lesions, including direct repair, NER, base excision repair, inter-strand cross-link (ICL) repair and double-strand break (DSB) repair (39). HELQ maintains genomic stability and avoids tumorigenesis through its involvement in different repair pathways, including NER, DSB repair and ICL repair (9,1113).

A NER pathway is involved in the removal of DNA damage after certain types of DNA damage have occurred (13). The expression levels of NER pathway proteins [e.g., xeroderma pigmentosum complementation group A (XPA), XPC, replication protein A (RPA) and ERCC excision repair 1, endonuclease non-catalytic subunit] are important mediators for responses to platinum-based chemotherapy and influence DNA repair activity (13,42). HELQ is crucial for cellular responses to cisplatin through its role in regulating the expression of NER pathway proteins (13).

DSBs are major DNA lesions deleterious to cell survival and genetic stability (43). When DSBs are not repaired, they can result in chromosome loss and rearrangements and even carcinogenesis (44). Two major pathways can repair DSBs: Homologous recombination (HR) and non-homologous end joining (NHEJ) (45,46), as well as alternative pathways, such as microhomology-mediated end joining (MMEJ) and single strand annealing (SSA) (45,46). HR is essential for DSB repair in post-replicative chromatin following replication fork collapse (47,48), and requires the loading of the RAD51 recombinase onto single-stranded DNA (ssDNA) through DNA ends or at post-replicative ssDNA gaps (45). The function of HELQ in HR is to capture RPA-bound ssDNA and then to displace it to speed up the annealing of complementary DNA strands (9). HELQ immediately interacts with BCDX2, a paralog of RAD51, to accelerate effective HR in damaged replication forks (11). RFS-1 binds to HELQ and plays a complementary role in facilitating the breakdown of RAD51 dsDNA filaments in postsynaptic HR intermediates, which is required to complete meiotic DSB repair in Caenorhabditis elegans (47). In addition, the HR factor with OB-fold (HROB)-mini-chromosome maintenance 8 (MCM8)-MCM9 pathway functions redundantly with HELQ, supporting a postsynaptic step of HR in a parallel pathway (49). HELQ accelerates HR-efficiency at compromised replication forks by working in parallel with the FA pathway in HELQΔC/ΔC and FANCD2−/− double mutant mice (11). Furthermore, HELQ is essential for the function of the synthesis-dependent strand annealing modes of HR, MMEJ of G4-induced DSBs and SSA in genome stability and tumor avoidance (9).

ICLs are also deleterious DNA lesions caused by endogenous (malondialdehyde) or exogenous [mitomycin C (MMC); cis-platinum and psoralens] sources (50,51), which induce mutations and chromosomal rearrangements by inhibiting DNA replication and transcription (1,34). ICL repair is complex and proteins from the NER, translation synthesis and HR pathways are involved in ICL repair (40). With its association with RAD51 paralogs, HELQ prevents germ cell loss and reduces cancer susceptibility through ICL repair (11,52). HELQ deficiency leads to germ cell attrition and ICL repair sensitivity in mouse and human cells (11,12,53). The FA pathway plays a critical role in recruiting RAD51-mediated HR during ICL repair (40). In parallel with HELQ, HROB participates in ICL repair as epistatic with MCM8-MCM9 (49), which also directly facilitates the repair of ICL-induced damage downstream of FANCD2 ubiquitylation (49,54).

DNA binding and unwinding

The HELQ HTH domain is associated with DNA strand binding and protein interactions (2,34). The dissociation constant values of wild-type HELQ for ssDNA and dsDNA were reported as 0.14 and 5.3 µM, respectively, indicating a strong preference for ssDNA binding and suggesting that the protein must track along, displacing the dsDNA strand (2). Furthermore, HELQ mutations reduce dsDNA binding, but ssDNA binding is not affected (55). The HELQ-ssDNA interaction is essential for the translocation mechanism. Mechanistically, HELQ interacts with RPA, and RPA coordinates the loading of HELQ onto ssDNA. HELQ helicase core is activated by ATP-Mg2+ binding and translocates along ssDNA as a dimer when loaded onto ssDNA (35).

In addition, HELQ is an ssDNA-activated ATPase, which is important for unwinding forked DNA (10,56). As well as unwinding ssDNA and dsDNA junctions, and HELQ is capable of unwinding 3′ overhangs, 3′ lagging strand forks, Y-structures, and D-loops (3); however, HELQ cannot unwind using ATPγS or 5′ overhang substrates (9). The HELQ unwinding of 3′ overhang substrates is inhibited by RPA, whereas RAD51 stimulates the unwinding activity of its D-loops by forming a complex with HELQ (9).

DNA replication

HELQ is an ATP-dependent enzyme involved in the recovery of replication forks that have stalled or collapsed following DNA damage (3). HELQ can act on damaged replication forks where the leading strand template for DNA replication has stopped, causing the polymerase to uncouple and continue DNA synthesis from the lagging strand template (10). Following treatment with camptothecin (CPT), an agent that stalls and collapses replication forks, HELQ is recruited to stalled replication sites that are associated with replication resumption. HELQ can facilitate replication resumption, possibly through colocalizing with IdU incorporation sites and RPA foci, by unwinding the nascent lagging strand, or alternatively through HELQ co-localization with RAD51 and FANCD2 at sites of stalled replication (3,12). In addition, HELQ has an important role in rescuing stalled forks during the normal S phase, and the loss of HELQ results in increased stalled forks, a role which is not epistatic with that of FA complementation group C (53).

DNA strand annealing

Previous studies of HELQ have focused more on its DNA repair and DNA unwinding functions, while its role in DNA strand annealing has been underappreciated. Tafel et al (3) reported that HELQ does not exhibit a strong annealing activity, and that RPA can suppress separated strand reannealing by binding to unwound ssDNA generated by HELQ. By contrast, Anand et al (9) reported that RPA strongly accelerated the DNA strand annealing activity of HELQ. Mechanistically, in addition to capturing RPA-bound DNA strands, HELQ is capable of superseding RPA and stimulating complementary DNA strands. Furthermore, it was found that ATP binding and hydrolysis are essential for HELQ DNA annealing activity, whereas the RAD51 addition was unaffected by HELQ-dependent DNA annealing activity (9).

Gene functions regulated by HELQ

One important question is ‘How are HELQ, its downstream mediators, and their mechanisms related in normal and cancer cells?’ Further, there are various studies reporting that HELQ can contribute to both germ cell maintenance and tumorigenesis in mammals by regulating its target genes: CHK1, FANCD2, RAD51 and RPA (3,4,9,14).

CHK1

CHK1 is a primary effector of DNA lesion and replication checkpoint responses, and its inhibition promotes DNA damage and reduces HR repair (57,58). CHK1 physically interacts with RAD51, while CHK1/RAD51 disruption or inactivation induces defective HR, aberrant replication dynamics, and chromosome instability (59). In osteosarcoma cells, CHK1 activation is promoted by HELQ, and HELQ colocalizes with RAD51 to participate in the repair of damaged forks by HR (3).

FANCD2

FA is a rare genetic disease, and patients with FA are hypersensitive to DNA ICL-inducing agents such as mitomycin C and cisplatin (60). FA signal transduction involves 22 proteins, which share a common pathway that is activated on DNA damage (61). FANCD2 is currently the focus of research into the FA pathway, and is crucial in cellular responses to DNA lesions (62). HR may be regulated by the FA pathway, where activated FANCD2 is translocated to chromatin-associated foci, where it colocalizes with HR proteins [RAD51 and breast invasive carcinoma (BRCA)2] (63). The strong negative effect of HELQ knockdown on single-stranded templated repair via the HR repair pathway may be explained by its physical interaction with FANCD2 and RAD51 (64). Following CPT treatment, HELQ is involved in fork repair and restart by localizing to replication forks, unwinding lagging strand structures, and co-localizing with RAD51 and FANCD2 (3,14). In addition, although the FA pathway, ICL sensitivities, and HELQ are additive, there is no epistasis of HELQ for downstream targets of FANCD2, and HELQ plays an independent role from FA pathway in ICL processing (12).

RAD51

RAD51 is a DNA-binding protein that can bind to both ssDNA and dsDNA and maintain genome stability in DNA replication (65,66). RAD51 is also an ATPase that forms nucleoprotein filaments on ssDNA and facilitates the search for homologous repair templates, such as homologous chromosomes, sister chromatids or ectopic homologous sequences (67). Hence, RAD51 is essential for regulating fork reversal by discovering and invading homologous DNA sequences during DSB repair by HR (65,67). In addition, five RAD51 paralogs [RAD51B/C/D and X-ray repair cross complementing (XRCC)2/3] that form two main complexes, BCDX2 (RAD51B/C/D-XRCC2) and CX3 (RAD51C-XRCC3), are involved in the HR repair of collapsed replication forks and maintenance of genome stability (6870). The BCDX2 complex preferentially binds to ssDNA and accelerates HELQ ATPase activity (71), which plays a central role in recognizing damage and stimulating fork remodeling in response to fluctuating dNTP pools (72). Adelman et al (11) found that HELQ plays a critical role in replication-coupled HR by interacting with complexes of the RAD51 paralog, BCDX2, to avoid germ cell loss and tumorigenesis. Anand et al (9) also demonstrated that HELQ binds to BCDX2 complexes, and that the latter strongly stimulates the translocation of HELQ during DNA unwinding. Takata et al (12) reported that HELQ interacts with ATR and HR-related RAD51 paralogs and participates in DNA LCL resistance in human cells.

RPA

As a major ssDNA binding protein, RPA is abundant in eukaryotic cells (66,73). RPA comprises three subunits, RPA14, RPA32 and RPA70, which are first responders in the event of the disruption of DNA metabolism. RPA is essential for replication, recombination and repair by binding to ssDNA to maintain genome duplication and stability (74). RPA and HELQ physically interact to form a supershift complex with HELQ on ssDNA (4). Furthermore, RPA stimulates HELQ helicase activity by binding to unwound regions generated by HELQ and inhibiting reannealing (4). Similarly, in mammals, RPA can restrain DNA unwinding while promoting DNA strand annealing by HELQ. Mechanistically, HELQ is recruited to ssDNA by interacting with RPA, followed by RPA displacement to stimulate complementary DNA strand annealing (9).

The roles of HELQ in reproduction

HELQ was first identified in the mouse and human genomes through its homology to D. melanogaster Mus308 (1). HELQ is expressed at different levels in various tissues, including the testes, ovaries, skeletal muscle and heart (75) in humans, and has also been detected in several other tissues (https://www.proteinatlas.org/ENSG00000163312-HELQ/tissue) (Fig. 2).

HELQ plays a crucial role in germ cell maintenance, and its loss results in subfertility. A high level of HELQ expression is found in the reproductive system, including the ovaries, testes, cervix, breast, epididymis, endometrium and prostate. In HELQ-deficient mice, HELQ facilitates RAD51 paralog-dependent repair, thereby preventing germ cell attrition (11). It is disputed whether mutant HELQ has an adverse effect on the male reproductive system. HELQgt/gt male mice have significantly smaller testes, suffer from seminiferous tubule atrophy, and lack spermatocytes and spermatogonia, as compared with wild-type male mice, indicating that HELQ deficiency leads to a mild form of hypogonadism (53); however, two heterozygous mutant mouse models (HELQ+/M5 and HELQ+/M6) did not show any spermatogenic defects, indicating that heterozygous HELQ variants alone do not cause development of the sertoli cell-only syndrome phenotype in mice (76). Furthermore, Wang et al (77) found no evidence that HELQ mutations are associated with premature ovarian failure in Chinese women.

Potential roles of HELQ in tumorigenesis and underlying mechanisms

In addition to its physiological functions, HELQ is often implicated in tumorigenesis. Genetic alterations, such as deletions and mismatches, are frequently associated with tumorigenesis. Single-nucleotide polymorphism variants of HELQ have been associated with an increased risk of various cancers, including upper aerodigestive tract cancers (78,79), esophageal squamous cell carcinoma (80,81), head and neck cancers (82), gastric adenocarcinoma (81), and breast and ovarian cancer (OV) (8385) through genome-wide association studies (Table III). Furthermore, there is accumulating evidence indicating that HELQ may act as a tumor suppressor for several cancers, such as osteosarcoma (86), OV (12,13,34,77,87), chronic lymphocytic leukemia (CLL) (88), non-small cell lung cancer (NSCLC) (89) and endometrial stromal sarcoma (ESS) (Table IV) (90). HELQ is an important regulator of cancer proliferation, invasion, migration and can contribute to poor patient prognosis and platinum resistance through several mechanisms (13,86,87) in different types of tumors (Fig. 3).

Table III.

Associations between SNPs within HELQ and cancer risk.

Table III.

Associations between SNPs within HELQ and cancer risk.

Alleles

SNPGenotypeMajorMinorReason for replication attemptCancer typeRisk factorsSignificant association(Refs.)
rs1494961/TCNon-synonymousUpper aero-digestive tract (UADT) cancersAge and sexYes, P=1×10−8(78)
rs1494961//CMissense mutation V306I Smoking and/or alcoholYes, P=2.65×10−4(79)
rs1494961C/C + C/T and T/T///Head and neck squamous cell carcinomaAlcohol consumption and smoking pack-yearsYes, P<0.001(82)
rs1494961TT, TC, CCTC/ESCCAge, Smoking, DrinkingYes, P=0.032(80)
rs13115704/TC/ESCC/Yes, P=8.07×10−3(81)
rs1494961/TC/Gastric cancer (GC)/Yes, P=0.035
rs13141136///Synonymous andBreast and/No, P>0.05(83)
rs7665103 non-synonymousovarian cancer
rs141700135 missense variants
rs138939487
rs1494961
Rs11099601 AG/Breast cancer/Yes, P=5.62×10−6(84)
Rs4693089GG GA/AA G/Breast cancer/Yes, P=2.38×10−19(85)

[i] ESCC, esophageal squamous cell carcinoma.

Table IV.

Summary of the current literature on HELQ dysregulation in different cancers.

Table IV.

Summary of the current literature on HELQ dysregulation in different cancers.

CancerDeregulationDownstream targetsPhenotypic effectStudies(Refs.)
OsteosarcomaLowCHK1↓, RAD51↓DNA repair↓, cell invasion↑, cell migration↑, cell proliferation↑In vitro(86)
CLLLow (CLL vs. normal B cells)MYC↓, E2F1↓, DNA repair pathway↓, Hedgehog signaling↑, Kras signaling pathway↑response to immuno-chemotherapy ↓, Richter transformation↓, unfavorable OSGEO dataset(88)
Ovarian cancerLoss/mutationRAD51B/C/D↓, XRCC2↓, CHK1↓G2/M arrest↓, cellular sensitivity↑, chromosome radial formation↑In vitro(12)
High-grade serous ovarian cancerHigh in platinum resistanceXAB2↑Poor prognosis, DNA damage repair↑, platinum resistance↑, apoptosis↓In vitro and TCGA database(87)
Cisplatin-resistant ovarian cancerHigh in cisplatin-resistantNER pathway proteins (RPA32, RAP70, XPA, XPC, ERCC1)↑, ATM/ATR pathway protein↑The cellular resistance to cisplation↑, DNA repair activity↑, Nucleotide excision repair pathway↑In vitro(13)
Non-small cell lung cancerLowRIPK3↓Cell proliferation↑, cell migration↑, chemotactic response↑, DNA damage repair↓, cell necrosis↓, cell death↓In vitro and in vivo(89)
Endometrial stromal sarcomaLowRAD51C↓DNA repair↓Clinical tissues(90)

[i] CLL, chronic lymphocytic leukemia.

Osteosarcoma

HELQ plays a critical role in tumor suppression in mammals through interacting with the BCDX2 complex (11). HELQ overexpression inhibited osteosarcoma cell proliferation, migration, invasion and DNA damage repair (86). In addition, the antitumor activity of HELQ may be associated with the upregulation of the DNA damage-related proteins RAD51 and CHK1expression, and HELQ modulates an anti-invasive phenotype and DNA damage repair in osteosarcoma cells by activation of the CHK1-RAD51 signaling pathway (86).

NSCLC

HELQ is downregulated in NSCLC tissues and cells, while the malignancy of lung cancer cells was enhanced by HELQ depletion. HELQ overexpression inhibits cell proliferation and migration by suppressing DNA damage repair, and promotes cell death by inducing necrosis through its interaction with receptor-interacting serine/threonine kinase 3 (RIPK3) (89). HELQ is a favorable prognostic factor for patients with NSCLC, and low HELQ levels in patients with NSCLC were associated with a reduced overall survival (OS) through a Kaplan-Meier plotter.

ESS

HELQ and RAD51C expression levels are decreased in ESS compared with normal endometrial tissues. HELQ expression was found to be correlated with the size and type of ESS. Neither HELQ nor RAD51C expression were correlated with age, FIGO stage or lymph node metastasis status. The occurrence and development of ESS may be affected by DNA repair involving HELQ and RAD51C (90).

OV

In addition, HELQ plays a role in cellular resistance in response to ICLs and G2/M arrest, through its association with the RAD51 paralogs RAD51B/C/D and XRCC2, and promotes the activation of the ATR substrate, CHK1 (12). HELQ and XPA binding protein 2 are associated with platinum resistance, poor prognosis, decreased apoptosis and increased DNA damage repair in ascites from high-grade serous OV cells (87). In addition, HELQ decreases cisplatin sensitivity in OV cells by activating the NER and ATM/ATR pathways (13). OV patients with HELQ-low expression had improved overall and disease-free survival than those with a high HELQ level. There is evidence that HELQ can be used independently as a prognostic marker to predict survival in patients with OV (13).

CLL

CLL patients with low HELQ levels exhibited a significantly unfavorable OS compared with patients with high HELQ levels. HELQ may be useful in predicting patients at high risk for CLL based on the Richter transformation. Higher HELQ expression was also associated with an improved response to immuno-chemotherapy in patients with CLL. HELQ represented a prognostic marker for CLL associated with the activation of MYC signaling, E2 factor signaling and DNA repair pathways, as well as the suppression of Hedgehog and Kras signaling (88).

Pan-cancer analysis of HELQ

Pan-cancer analysis of HELQ was conducted using The Cancer Genome Atlas (TCGA) (https://tcga-data.nci.nih.gov/tcga/) and Genotype-Tissue Expression (GTEx) project (https://gtexportal.org/) datasets. In TCGA dataset, HELQ expression was significantly lower in bladder urothelial carcinoma (BLCA), BRCA, cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), glioblastoma multiforme (GBM), colon adenocarcinoma (COAD), kidney chromophobe (KICH), kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP), lung squamous cell carcinoma (LUSC), lung adenocarcinoma (LUAD), pancreatic adenocarcinoma (PAAD), pheochromocytoma and paraganglioma (PCPG), rectum adenocarcinoma (READ), prostate adenocarcinoma (PRAD), stomach adenocarcinoma (STAD), thyroid carcinoma (THCA), and uterine corpus endometrial carcinoma (UCEC) than that in normal tissues, while HELQ expression in cholangiocarcinoma (CHOL), esophageal carcinoma (ESCA), head and neck squamous cell carcinoma (HNSC) and liver hepatocellular carcinoma (LIHC) were significantly higher (Fig. 4A). In combined TCGA and GTEx datasets, HELQ expression levels in adrenocortical carcinoma, BRCA, BLCA, COAD, CESC, ESCA, KIRC, KICH, KIRP, LUSC, LUAD, PAAD, OV, PRAD, PCPG, READ, THCA, testicular germ cell tumors, uterine carcinosarcoma and UCEC were significantly lower than those in normal tissues, while in CHOL, GBM, lymphoid neoplasm diffuse large B-cell lymphoma, HNSC, LIHC, brain lower grade glioma, PAAD, STAD, skin cutaneous melanoma and thymoma were significantly higher (Fig. 4B).

Figure 4.

Pan-cancer analysis of HELQ expression in (A) TCGA dataset and (B) TCGA + GTEx combined dataset. (A) The expression of HELQ in BLCA, BRCA, CESC, GBM, COAD, KICH, KIRC, KIRP, LUSC, LUAD, PAAD, PCPG, READ, PRAD, STAD, THCA and UCEC cancer types was significantly lower than that in normal tissues, while levels in CHOL, ESCA, HNSC and LIHC were significantly higher in TCGA dataset. (B) HELQ expression levels in ACC, BRCA, BLCA, COAD, CESC, ESCA, KIRC, KICH, KIRP, LUSC, LUAD, PAAD, OV, PRAD, PCPG, READ, THCA, TGCT, UCS and UCEC were significantly lower than those in normal tissues, while levels in CHOL, GBM, DLBC, HNSC, LIHC, LGG, PAAD, STAD, SKCM and THYM were significantly higher in combined TCGA and GTEx datasets. HELQ, helicase POLQ-like; TCGA, The Cancer Genome Atlas; GTEx, BLCA, bladder urothelial carcinoma; BRCA: breast invasive carcinoma; CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma; GBM, glioblastoma multiforme; COAD, colon adenocarcinoma; KICH, kidney chromophobe; KIRC, kidney renal clear cell carcinoma; KIRP, kidney renal papillary cell carcinoma; LUSC, lung squamous cell carcinoma; LUAD, lung adenocarcinoma; PAAD, pancreatic adenocarcinoma; PCPG, pheochromocytoma and paraganglioma; READ, rectum adenocarcinoma; PRAD: prostate adenocarcinoma; STAD, stomach adenocarcinoma; THCA: thyroid carcinoma; UCEC, uterine carcinosarcoma; CHOL: cholangiocarcinoma; ESCA, esophageal carcinoma; HNSC, head and neck squamous cell carcinoma; LIHC, liver hepatocellular carcinoma; OV, ovarian serous cystadenocarcinoma; TGCT, testicular germ cell tumors; DLBC, lymphoid neoplasm diffuse large B-cell lymphoma; SKCM: skin cutaneous melanoma; THYM, thymoma. *P<0.05, **P<0.01, ***P<0.001.

Targeting HELQ for potential treatment

HELQ, a superfamily II DNA helicase, is a tumor suppressor that can mediate tumor-inhibiting activity. Furthermore, HELQ is pivotal in tumor proliferation, metastasis, platinum resistance, cell-cycle checkpoint regulation and DNA damage response. These important roles of HELQ in tumorigenesis highlight its potential as a target for the development of novel cancer therapeutics. Nevertheless, a few small compounds or drugs that target HELQ have been identified for potential treatment (Table V). Kras signaling is negatively associated with HELQ expression in patients with CLL, and ulixertinib, a Kras signaling inhibitor, may offer a new therapeutic option for patients with HELQ-low CLL (88,91). Furthermore, topotecan, a topoisomerase I inhibitor, is sensitive in HELQ mutants that induce single-ended DNA DSBs in replicating cells (92). In additionally, a RAD51 inhibitor, Cam833, which disrupts the interaction between RAD51 and BRCA2, synergizes with the poly (ADP-ribose) polymerase (PARP) inhibitors (93). HR efficacy was reduced by 50–60% in HELQ knockout mice with increased sensitivity to PARP inhibitors (11). It was hypothesized that PARP inhibitors may also play a role in patients with HELQ deficiency, but further studies are required to test this hypothesis. HELQ-deficient mice and cells show hypersensitivity to the ICL agent MMC through more chromatid breaks and radial chromosomes (11). HELQ-deficient cells were also hypersensitive to CPT, a replication inhibiting agent (11).

Table V.

Description of HELQ as therapeutic target at present.

Table V.

Description of HELQ as therapeutic target at present.

Compound Drugs/InhibitorsTarget HELQMechanism(Refs.)
UlixertinibA Kras signaling inhibitorHELQ-low CLLInhibit the Kras signaling, negatively associated HELQ(88)
TopotecanA topoisomerase I inhibitorHELQ mutantsinduce one-ended DNA DSBs(92)
Cam833A RAD51 inhibitorHELQ deficiencyPrevent RAD51-mediated homologous recombination(93)
PARPiA PARP inhibitorHELQ deficiencyNA(11)
Mitomycin CICL agentHELQ deficiencyInhibit replication(11)
CamptothecinReplication blocking agentHELQ deficiencyInhibit replication(11)

[i] HELQ, helicase POLQ-like.

Conclusions

HELQ is a DNA helicase with multiple biological functions under normal and pathological conditions. The diverse functions of HELQ, including DNA binding, DNA unwinding and DNA repair, among others, involve three HELQ protein domains: a DEAD/DEAH box helicase domain, a helicase C-terminal domain, and a HTH (HTH_61) domain. The DEAD/DEAH box helicase domain is responsible for ATP binding-mediated RNA metabolism, the helicase C-terminal domain is involved in ATP-dependent DNA or RNA unwinding, and the HTH domain is important for DNA strand binding and protein interactions. Under normal conditions, HELQ is abundantly expressed in healthy human tissues, especially in the reproductive system, where it plays an important role in maintaining germ cell viability through RAD51 paralog-dependent repair. In addition, recent studies have reported that HELQ is abnormally expressed in various cancers (8690). HELQ is important for cell proliferation, metastasis and chemotherapy resistance by regulating various molecular signaling pathways (for example, the NER, CHK1-RAD51 and ATM/ATR pathways). HELQ deficiency is also associated with the clinical characteristics and prognosis of patients with cancer, and is considered a novel prognostic biomarker and potentially critical target in cancer therapy.

Of note, multiple biological processes involving HELQ are associated with tumorigenesis, and the protein plays a particular role in the reproductive system. Therefore, further studies focusing on the physiological roles of HELQ, as well as the detailed mechanisms underlying the relationship between HELQ and tumor occurrence under different pathological conditions, are warranted.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81902802), the National Science Foundation of Guangdong (grant no. 2019A1515010633), the Guangzhou Science and Technology Project (grant nos. 202102010060, 202102020766 and 202201010020), and the Guangzhou Science and Technology Project of Traditional Chinese Medicine and Integrated Traditional and Western Medicine (grant no. 20232A011007).

Availability of data and materials

Not applicable.

Authors' contributions

XX and NT wrote original draft preparation. NT and WW prepared figures and tables; ZL and YW wrote review and revision; NT, XX, ZL and YW were responsible for conceptual design; YW carried out supervision and management. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

ATM

ataxia telangiectasia mutated protein

ATR

ataxia telangiectasia and Rad3-related protein

BLCA

bladder urothelial carcinoma

BRCA

breast invasive carcinoma

CESC

cervical squamous cell carcinoma and endocervical adenocarcinoma

CHK1

checkpoint kinase 1

CHOL

cholangiocarcinoma

COAD

colon adenocarcinoma

CPT

camptothecin

dsDNA

double-stranded DNA

DSBs

DNA double-stranded breaks

ESCA

esophageal carcinoma

ESS

endometrial stromal sarcoma

FA

Fanconi anemia

FANCD2

Fanconi anemia group D2

GBM

glioblastoma multiforme

GTEx

genotype-tissue expression

HELQ

helicase POLQ-like

HelicC

helicase C-terminal domain

HNSC

head and neck squamous cell carcinoma

HR

homologous recombination

HROB

homologous recombination factor with OB-fold

HTH

helix-turn-helix

ICL

inter-strand cross-link

KICH

kidney chromophobe

KIRC

kidney renal clear cell carcinoma

KIRP

kidney renal papillary cell carcinoma

LIHC

liver hepatocellular carcinoma

LUAD

lung adenocarcinoma

LUSC

lung squamous cell carcinoma

MCM

mini-chromosome maintenance

MMEJ

microhomology mediated end joining

NER

nucleotide excision repair

NHEJ

non-homologous end joining

OS

overall survival

OV

ovarian cancer

PAAD

pancreatic adenocarcinoma

PARP

poly ADP ribose polymerase

PCPG

pheochromocytoma and paraganglioma

PRAD

prostate adenocarcinoma

RAD51

RecA-like ATP-dependent recombinase 51

READ

rectum adenocarcinoma

RIPK3

receptor-interacting serine/threonine kinase 3

RPA

replication protein A

SF2

superfamily-2

SSA

single strand annealing

STAD

stomach adenocarcinoma

ssDNA

single-stranded DNA

TCGA

The Cancer Genome Atlas

THCA

thyroid carcinoma

UCEC

uterine corpus endometrial carcinoma

References

1 

Marini F and Wood RD: A human DNA helicase homologous to the DNA cross-link sensitivity protein Mus308. J Biol Chem. 277:8716–8723. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Richards JD, Johnson KA, Liu H, McRobbie AM, McMahon S, Oke M, Carter L, Naismith JH and White MF: Structure of the DNA repair helicase hel308 reveals DNA binding and autoinhibitory domains. J Biol Chem. 283:5118–5126. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Tafel AA, Wu L and McHugh PJ: Human HEL308 localizes to damaged replication forks and unwinds lagging strand structures. J Biol Chem. 286:15832–15840. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Woodman IL and Bolt EL: Winged helix domains with unknown function in Hel308 and related helicases. Biochem Soc Trans. 39:140–144. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Singleton MR, Dillingham MS and Wigley DB: Structure and mechanism of helicases and nucleic acid translocases. Ann Rev Biochem. 76:23–50. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Fairman-Williams ME, Guenther UP and Jankowsky E: SF1 and SF2 helicases: Family matters. Curr Opin Struct Biol. 20:313–324. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Bleichert F and Baserga SJ: The long unwinding road of RNA helicases. Mol Cell. 27:339–352. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Woodman IL, Briggs GS and Bolt EL: Archaeal Hel308 domain V couples DNA binding to ATP hydrolysis and positions DNA for unwinding over the helicase ratchet. J Mol Biol. 374:1139–1144. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Anand R, Buechelmaier E, Belan O, Newton M, Vancevska A, Kaczmarczyk A, Takaki T, Rueda DS, Powell SN and Boulton SJ: HELQ is a dual-function DSB repair enzyme modulated by RPA and RAD51. Nature. 601:268–273. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Guy CP and Bolt EL: Archaeal Hel308 helicase targets replication forks in vivo and in vitro and unwinds lagging strands. Nucleic Acids Res. 33:3678–3690. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Adelman CA, Lolo RL, Birkbak NJ, Murina O, Matsuzaki K, Horejsi Z, Parmar K, Borel V, Skehel JM, Stamp G, et al: HELQ promotes RAD51 paralogue-dependent repair to avert germ cell loss and tumorigenesis. Nature. 502:381–384. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Takata K, Reh S, Tomida J, Person MD and Wood RD: Human DNA helicase HELQ participates in DNA interstrand crosslink tolerance with ATR and RAD51 paralogs. Nat Commun. 4:23382013. View Article : Google Scholar : PubMed/NCBI

13 

Long J, Zhu JY, Liu YB, Fu K, Tian Y, Li PY, Yang WQ, Yang SY, Yin JY, Yin G and Zhang Y: Helicase POLQ-like (HELQ) as a novel indicator of platinum-based chemoresistance for epithelial ovarian cancer. Gynecol Oncol. 149:341–349. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Song X, Ni J and Shen Y: Structure-based genetic analysis of Hel308a in the Hyperthermophilic Archaeon Sulfolobus islandicus. J Genet Genomics. 43:405–413. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Gorbalenya AE, Koonin EV, Donchenko AP and Blinov VM: Two related superfamilies of putative helicases involved in replication, recombination, repair and expression of DNA and RNA genomes. Nucleic Acids Res. 17:4713–4730. 1989. View Article : Google Scholar : PubMed/NCBI

16 

Caruthers JM and McKay DB: Helicase structure and mechanism. Curr Opin Struct Biol. 12:123–133. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Pyle AM: Translocation and unwinding mechanisms of RNA and DNA helicases. Ann Rev Biophys. 37:317–336. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Jankowsky E: RNA helicases at work: Binding and rearranging. Trends Biochem Sci. 36:19–29. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Johnson SJ and Jackson RN: Ski2-like RNA helicase structures: Common themes and complex assemblies. RNA Biol. 10:33–43. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Bennett RJ and Keck JL: Structure and function of RecQ DNA helicases. Crit Rev Biochem Mol Biol. 39:79–97. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Fuller-Pace FV: DExD/H box RNA helicases: Multifunctional proteins with important roles in transcriptional regulation. Nucleic Acids Res. 34:4206–4215. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Lusser A and Kadonaga JT: Chromatin remodeling by ATP-dependent molecular machines. Bioessays. 25:1192–1200. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Jankowsky E and Fairman ME: RNA helicases-one fold for many functions. Curr Opin Struct Biol. 17:316–324. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Jankowsky E and Bowers H: Remodeling of ribonucleoprotein complexes with DExH/D RNA helicases. Nucleic Acids Res. 34:4181–4188. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Byrd AK and Raney KD: Superfamily 2 helicases. Front Bioscience. 17:2070–2088. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Han X, Zhao L and Li X: HELQ in cancer and reproduction. Neoplasma. 63:825–835. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Cordin O, Banroques J, Tanner NK and Linder P: The DEAD-box protein family of RNA helicases. Gene. 367:17–37. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Heung LJ and Del Poeta M: Unlocking the DEAD-box: A key to cryptococcal virulence? J Clin Invest. 115:593–595. 2005. View Article : Google Scholar : PubMed/NCBI

29 

de la Cruz J, Kressler D and Linder P: Unwinding RNA in Saccharomyces cerevisiae: DEAD-box proteins and related families. Trends Biochem Sci. 24:192–198. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Aubourg S, Kreis M and Lecharny A: The DEAD box RNA helicase family in Arabidopsis thaliana. Nucleic Acids Res. 27:628–636. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Tanner NK and Linder P: DExD/H box RNA helicases: From generic motors to specific dissociation functions. Mol Cell. 8:251–262. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Huffman JL and Brennan RG: Prokaryotic transcription regulators: More than just the helix-turn-helix motif. Curr Opin Struct Biol. 12:98–106. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Kitano K, Kim SY and Hakoshima T: Structural basis for DNA strand separation by the unconventional winged-helix domain of RecQ helicase WRN. Structure. 18:177–187. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Li YP, Yang JJ, Xu H, Guo EY and Yu Y: Structure-function analysis of DNA helicase HELQ: A new diagnostic marker in ovarian cancer. Oncol Lett. 12:4439–4444. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Jenkins T, Northall SJ, Ptchelkine D, Lever R, Cubbon A, Betts H, Taresco V, Cooper CDO, McHugh PJ, Soultanas P and Bolt EL: The HelQ human DNA repair helicase utilizes a PWI-like domain for DNA loading through interaction with RPA, triggering DNA unwinding by the HelQ helicase core. NAR Cancer. 3:zcaa0432021. View Article : Google Scholar : PubMed/NCBI

36 

Gaudet P, Livstone MS, Lewis SE and Thomas PD: Phylogenetic-based propagation of functional annotations within the Gene Ontology consortium. Brief Bioinform. 12:449–462. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Moldovan GL, Madhavan MV, Mirchandani KD, McCaffrey RM, Vinciguerra P and D'Andrea AD: DNA polymerase POLN participates in cross-link repair and homologous recombination. Mol Cell Biol. 30:1088–1096. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Wu L and Hickson ID: DNA helicases required for homologous recombination and repair of damaged replication forks. Ann Rev Genet. 40:279–306. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K and Linn S: Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Ann Rev Biochem. 73:39–85. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Li X and Heyer WD: Homologous recombination in DNA repair and DNA damage tolerance. Cell Res. 18:99–113. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Roy S: Maintenance of genome stability in plants: Repairing DNA double strand breaks and chromatin structure stability. Front Plant Sci. 5:4872014. View Article : Google Scholar : PubMed/NCBI

42 

Martin LP, Hamilton TC and Schilder RJ: Platinum resistance: The role of DNA repair pathways. Clin Cancer Res. 14:1291–1295. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Kamp JA, Lemmens B, Romeijn RJ, Changoer SC, van Schendel R and Tijsterman M: Helicase Q promotes homology-driven DNA double-strand break repair and prevents tandem duplications. Nat Commun. 12:71262021. View Article : Google Scholar : PubMed/NCBI

44 

Hoeijmakers JH: Genome maintenance mechanisms for preventing cancer. Nature. 411:366–374. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Scully R, Panday A, Elango R and Willis NA: DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat Rev Mol Cell Biol. 20:698–714. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Gartner A and Engebrecht J: DNA repair, recombination, and damage signaling. Genetics. 220:iyab1782022. View Article : Google Scholar : PubMed/NCBI

47 

Ward JD, Muzzini DM, Petalcorin MI, Martinez-Perez E, Martin JS, Plevani P, Cassata G, Marini F and Boulton SJ: Overlapping mechanisms promote postsynaptic RAD-51 filament disassembly during meiotic double-strand break repair. Mol Cell. 37:259–272. 2010. View Article : Google Scholar : PubMed/NCBI

48 

San Filippo J, Sung P and Klein H: Mechanism of eukaryotic homologous recombination. Ann Rev Biochem. 77:229–257. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Hustedt N, Saito Y, Zimmermann M, Alvarez-Quilon A, Setiaputra D, Adam S, McEwan A, Yuan JY, Olivieri M, Zhao Y, et al: Control of homologous recombination by the HROB-MCM8-MCM9 pathway. Genes Dev. 33:1397–1415. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Niedernhofer LJ, Daniels JS, Rouzer CA, Greene RE and Marnett LJ: Malondialdehyde, a product of lipid peroxidation, is mutagenic in human cells. J Biol Chem. 278:31426–31433. 2003. View Article : Google Scholar : PubMed/NCBI

51 

McHugh PJ, Spanswick VJ and Hartley JA: Repair of DNA interstrand crosslinks: Molecular mechanisms and clinical relevance. Lancet Oncol. 2:483–490. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Adelman CA and Boulton SJ: Metabolism of postsynaptic recombination intermediates. FEBS Lett. 584:3709–3716. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Luebben SW, Kawabata T, Akre MK, Lee WL, Johnson CS, O'Sullivan MG and Shima N: Helq acts in parallel to Fancc to suppress replication-associated genome instability. Nucleic Acids Res. 41:10283–10297. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Garcia-Higuera I, Taniguchi T, Ganesan S, Meyn MS, Timmers C, Hejna J, Grompe M and D'Andrea AD: Interaction of the Fanconi anemia proteins and BRCA1 in a common pathway. Mol Cell. 7:249–262. 2001. View Article : Google Scholar : PubMed/NCBI

55 

Northall SJ, Buckley R, Jones N, Penedo JC, Soultanas P and Bolt EL: DNA binding and unwinding by Hel308 helicase requires dual functions of a winged helix domain. DNA Repair. 57:125–132. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Fujikane R, Shinagawa H and Ishino Y: The archaeal Hjm helicase has recQ-like functions, and may be involved in repair of stalled replication fork. Genes Cells. 11:99–110. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Smith J, Tho LM, Xu N and Gillespie DA: The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res. 108:73–112. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Montano R, Thompson R, Chung I, Hou H, Khan N and Eastman A: Sensitization of human cancer cells to gemcitabine by the Chk1 inhibitor MK-8776: Cell cycle perturbation and impact of administration schedule in vitro and in vivo. BMC Cancer. 13:6042013. View Article : Google Scholar : PubMed/NCBI

59 

Krajewska M, Fehrmann RS, Schoonen PM, Labib S, de Vries EG, Franke L and van Vugt MA: ATR inhibition preferentially targets homologous recombination-deficient tumor cells. Oncogene. 34:3474–3481. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Bagby GC Jr: Genetic basis of Fanconi anemia. Curr Opin Hematol. 10:68–76. 2003. View Article : Google Scholar : PubMed/NCBI

61 

Seki S, Ohzeki M, Uchida A, Hirano S, Matsushita N, Kitao H, Oda T, Yamashita T, Kashihara N, Tsubahara A, et al: A requirement of FancL and FancD2 monoubiquitination in DNA repair. Genes Cells. 12:299–310. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Nepal M, Che R, Ma C, Zhang J and Fei P: FANCD2 and DNA Damage. Int J Mol Sci. 18:18042017. View Article : Google Scholar : PubMed/NCBI

63 

Muzzini DM, Plevani P, Boulton SJ, Cassata G and Marini F: Caenorhabditis elegans POLQ-1 and HEL-308 function in two distinct DNA interstrand cross-link repair pathways. DNA Repair. 7:941–950. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Richardson CD, Kazane KR, Feng SJ, Zelin E, Bray NL, Schäfer AJ, Floor SN and Corn JE: CRISPR-Cas9 genome editing in human cells occurs via the Fanconi anemia pathway. Nat Genet. 50:1132–1139. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Zellweger R, Dalcher D, Mutreja K, Berti M, Schmid JA, Herrador R, Vindigni A and Lopes M: Rad51-mediated replication fork reversal is a global response to genotoxic treatments in human cells. J Cell Biol. 208:563–579. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Bhat KP and Cortez D: RPA and RAD51: Fork reversal, fork protection, and genome stability. Nat Struct Mol Biol. 25:446–453. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Bonilla B, Hengel SR, Grundy MK and Bernstein KA: RAD51 gene family structure and function. Ann Rev Genet. 54:25–46. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Somyajit K, Subramanya S and Nagaraju G: RAD51C: A novel cancer susceptibility gene is linked to Fanconi anemia and breast cancer. Carcinogenesis. 31:2031–2038. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Suwaki N, Klare K and Tarsounas M: RAD51 paralogs: Roles in DNA damage signalling, recombinational repair and tumorigenesis. Seminars Cell Dev Biol. 22:898–905. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Somyajit K, Saxena S, Babu S, Mishra A and Nagaraju G: Mammalian RAD51 paralogs protect nascent DNA at stalled forks and mediate replication restart. Nucleic Acids Res. 43:9835–9855. 2015.PubMed/NCBI

71 

Masson JY, Stasiak AZ, Stasiak A, Benson FE and West SC: Complex formation by the human RAD51C and XRCC3 recombination repair proteins. Proc Natl Acad Sci USA. 98:8440–8446. 2001. View Article : Google Scholar : PubMed/NCBI

72 

Rein HL, Bernstein KA and Baldock RA: RAD51 paralog function in replicative DNA Damage and tolerance. Curr Opin Genet Dev. 71:86–91. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Chen R and Wold MS: Replication protein A: Single-stranded DNA's first responder: Dynamic DNA-interactions allow replication protein A to direct single-strand DNA intermediates into different pathways for synthesis or repair. Bioessays. 36:1156–1161. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Oakley GG and Patrick SM: Replication protein A: Directing traffic at the intersection of replication and repair. Front Bioscience. 15:883–900. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Marini F, Kim N, Schuffert A and Wood RD: POLN, a nuclear PolA family DNA polymerase homologous to the DNA cross-link sensitivity protein Mus308. J Biol Chem. 278:32014–32019. 2003. View Article : Google Scholar : PubMed/NCBI

76 

Murtaza G, Yang L, Khan I, Unar A, Khan M, Huan Z, Khan R and Shi Q: Identification and functional investigation of novel heterozygous HELQ mutations in patients with Sertoli Cell-only Syndrome. Genet Test Mol Biomarkers. 25:654–659. 2021. View Article : Google Scholar : PubMed/NCBI

77 

Wang W, Zhao S, Zhuang L, Li W, Qin Y and Chen ZJ: The screening of HELQ gene in Chinese patients with premature ovarian failure. Reprod Biomed Online. 31:573–576. 2015. View Article : Google Scholar : PubMed/NCBI

78 

McKay JD, Truong T, Gaborieau V, Chabrier A, Chuang SC, Byrnes G, Zaridze D, Shangina O, Szeszenia-Dabrowska N, Lissowska J, et al: A genome-wide association study of upper aerodigestive tract cancers conducted within the INHANCE consortium. PLoS Genet. 7:e10013332011. View Article : Google Scholar : PubMed/NCBI

79 

Babron MC, Kazma R, Gaborieau V, McKay J, Brennan P, Sarasin A and Benhamou S: Genetic variants in DNA repair pathways and risk of upper aerodigestive tract cancers: Combined analysis of data from two genome-wide association studies in European populations. Carcinogenesis. 35:1523–1527. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Gao Y, He Y, Xu J, Xu L, Du J, Zhu C, Gu H, Ma H, Hu Z, Jin G, et al: Genetic variants at 4q21, 4q23 and 12q24 are associated with esophageal squamous cell carcinoma risk in a Chinese population. Hum Genet. 132:649–656. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Li WQ, Hu N, Hyland PL, Gao Y, Wang ZM, Yu K, Su H, Wang CY, Wang LM, Chanock SJ, et al: Genetic variants in DNA repair pathway genes and risk of esophageal squamous cell carcinoma and gastric adenocarcinoma in a Chinese population. Carcinogenesis. 34:1536–1542. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Liang C, Marsit CJ, Houseman EA, Butler R, Nelson HH, McClean MD and Kelsey KT: Gene-environment interactions of novel variants associated with head and neck cancer. Head Neck. 34:1111–1118. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Pelttari LM, Kinnunen L, Kiiski JI, Khan S, Blomqvist C, Aittomaki K and Nevanlinna H: Screening of HELQ in breast and ovarian cancer families. Fam Cancer. 15:19–23. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Hamdi Y, Soucy P, Adoue V, Michailidou K, Canisius S, Lemacon A, Droit A, Andrulis IL, Anton-Culver H, Arndt V, et al: Association of breast cancer risk with genetic variants showing differential allelic expression: Identification of a novel breast cancer susceptibility locus at 4q21. Oncotarget. 7:80140–80163. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Homer MV, Charo LM, Natarajan L, Haunschild C, Chung K, Mao JJ, DeMichele AM and Su HI: Genetic variants of age at menopause are not related to timing of ovarian failure in breast cancer survivors. Menopause. 24:663–668. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Liu DN, Zhou YF, Peng AF, Long XH, Chen XY, Liu ZL and Xia H: HELQ reverses the malignant phenotype of osteosarcoma cells via CHK1-RAD51 signaling pathway. Oncol Rep. 37:1107–1113. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Zhu F, Yang S, Lei M, He Q, Wu L and Zhang Y: DNA repair protein HELQ and XAB2 as chemoresponse and prognosis biomarkers in ascites tumor cells of high-grade serous ovarian cancer. J Oncol. 2022:75219342022. View Article : Google Scholar : PubMed/NCBI

88 

Guo C, Gao YY, Ju QQ, Zhang CX, Gong M and Li ZL: HELQ and EGR3 expression correlate with IGHV mutation status and prognosis in chronic lymphocytic leukemia. J Transl Med. 19:422021. View Article : Google Scholar : PubMed/NCBI

89 

Zhong NS, Tong WL, Zhang Y, Xiao SN, Liu JM, Li AA, Yao GL, Lin Q and Liu ZL: HELQ suppresses migration and proliferation of non-small cell lung cancer cells by repairing DNA damage and inducing necrosis. Cell Biol Int. 47:188–200. 2023. View Article : Google Scholar : PubMed/NCBI

90 

Liu Y, Zhang Y and Tian Y: Expressions of HELQ and RAD51C in endometrial stromal sarcoma and their clinical significance. Nan Fang Yi Ke Da Xue Xue Bao. 40:936–941. 2020.(In Chinese). PubMed/NCBI

91 

Gimenez N, Martinez-Trillos A, Montraveta A, Lopez-Guerra M, Rosich L, Nadeu F, Valero JG, Aymerich M, Magnano L, Rozman M, et al: Mutations in the RAS-BRAF-MAPK-ERK pathway define a specific subgroup of patients with adverse clinical features and provide new therapeutic options in chronic lymphocytic leukemia. Haematologica. 104:576–586. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Thomas A, Cox J, Wolfe KB, Mingalone CH, Yaspan HR and McVey M: Division of Labor by the HELQ, BLM, and FANCM Helicases during Homologous Recombination Repair in Drosophila melanogaster. Genes (Basel). 13:4742022. View Article : Google Scholar : PubMed/NCBI

93 

Scott DE, Francis-Newton NJ, Marsh ME, Coyne AG, Fischer G, Moschetti T, Bayly AR, Sharpe TD, Haas KT, Barber L, et al: A small-molecule inhibitor of the BRCA2-RAD51 interaction modulates RAD51 assembly and potentiates DNA damage-induced cell death. Cell Chem Biol. 28:835–847.e5. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2023
Volume 50 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tang N, Wen W, Liu Z, Xiong X and Wu Y: HELQ as a DNA helicase: Its novel role in normal cell function and tumorigenesis (Review). Oncol Rep 50: 220, 2023
APA
Tang, N., Wen, W., Liu, Z., Xiong, X., & Wu, Y. (2023). HELQ as a DNA helicase: Its novel role in normal cell function and tumorigenesis (Review). Oncology Reports, 50, 220. https://doi.org/10.3892/or.2023.8657
MLA
Tang, N., Wen, W., Liu, Z., Xiong, X., Wu, Y."HELQ as a DNA helicase: Its novel role in normal cell function and tumorigenesis (Review)". Oncology Reports 50.6 (2023): 220.
Chicago
Tang, N., Wen, W., Liu, Z., Xiong, X., Wu, Y."HELQ as a DNA helicase: Its novel role in normal cell function and tumorigenesis (Review)". Oncology Reports 50, no. 6 (2023): 220. https://doi.org/10.3892/or.2023.8657