Review
RTEL1: functions of a disease-associated helicase

https://doi.org/10.1016/j.tcb.2014.01.004Get rights and content

Highlights

  • RTEL1 is an Fe–S helicase that controls homologous recombination by dismantling D-loop intermediates.

  • RTEL1 maintains telomere integrity by disassembling T-loops and counteracting G4-DNA structures.

  • RTEL1 variants strongly associate with predisposition to glioma, astrocytoma, and glioblastoma.

  • Mutations in RTEL1 give rise to Hoyeraal–Hreidarsson syndrome.

DNA secondary structures that arise during DNA replication, repair, and recombination (3R) must be processed correctly to prevent genetic instability. Regulator of telomere length 1 (RTEL1) is an essential DNA helicase that disassembles a variety of DNA secondary structures to facilitate 3R processes and to maintain telomere integrity. The past few years have witnessed the emergence of RTEL1 variants that confer increased susceptibility to high-grade glioma, astrocytomas, and glioblastomas. Mutations in RTEL1 have also been implicated in Hoyeraal–Hreidarsson syndrome, a severe form of the bone-marrow failure and cancer predisposition disorder, dyskeratosis congenita. We review these recent findings and highlight its crucial link between DNA secondary-structure metabolism and human disease.

Section snippets

DNA secondary-structure metabolism during DNA replication and repair

The formation and/or metabolism of DNA secondary structures is important for many physiological processes, and is particularly relevant during DNA replication and repair (reviewed in [1]). However, persistent or aberrantly processed DNA secondary structures can have pathological consequences and are an established source of genome instability 2, 3. DNA secondary structures can form from alternative DNA sequence motifs [e.g., trinucleotide repeats, or G-rich DNA that forms four-stranded DNA

RTEL1 in telomere homeostasis

RTEL1 belongs to the DEAH (named from the corresponding four amino acid motif in single-letter code) subfamily of the superfamily 2 (SF2) helicases, which contain a RAD3-related DNA helicase domain with 5′ to 3′ helicase activity (reviewed in 19, 20). RTEL1 is also a member of the iron–sulfur (Fe–S) cluster helicase family, which includes xeroderma pigmentosum group D (XPD), Fanconi anemia complementation group J (FANCJ), and DEAD/H box helicase 11 (DDX11; reviewed in [21]). The activity of

RTEL1 controls recombination in mitotic and meiotic cells

RTEL1 was independently identified in C. elegans as a key regulator of HR in a genetic screen for synthetic lethality with mutation in the sgs1/BLM orthologue, which is associated with the accumulation of persistent recombination intermediates [28]. Consistent with a role for RTEL1 in suppressing HR (Figure 1), worms and human cells lacking RTEL1 exhibit hyper-recombination and sensitivity to DNA damaging agents. Moreover, C. elegans rtel-1 mutants are also synthetic lethal when combined with

RTEL1 controls recombination at telomeres

Visualization of vertebrate telomeres by electron microscopy and STORM (stochastic optical reconstruction microscopy) has revealed that some, if not all, chromosome ends adopt a lasso-like configuration called a T-loop 4, 33. The T-loop is proposed to form upon strand invasion of the 3′ ss TTAGGG telomeric repeats into an adjacent duplex of telomeric DNA, resulting in a D-loop intermediate at the site of strand invasion [34]. T-loops may protect the chromosome end from degradation and

RTEL1 is essential to facilitate replication

Several observations suggested a possible role for RTEL1 during DNA replication. Mouse ES cells deficient for RTEL1 exhibit reduced proliferative capacity, and worms and mammalian cells lacking RTEL1 are particularly sensitive to DNA damaging agents that hinder DNA replication, such as inter-strand crosslinking agents 25, 26, 28. Proteomic analysis of RTEL1 interacting proteins reinforced the link between RTEL1 and DNA replication, and identified multiple DNA replication factors, including

RTEL1 in human diseases

Mutations in XPD, FANCJ, and DDX11 have been shown to be an underlying cause of xeroderma pigmentosum (reviewed in [40]), Fanconi anemia 41, 42, 43, 44, and Warsaw breakage syndrome 45, 46, respectively. In the last few years, RTEL1 variants have also been linked to several distinct human brain cancers and recently, RTEL1 mutations have been shown to give rise to Hoyeraal–Hreidarsson syndrome, a severe form of the telomeropathy dyskeratosis congenita 14, 16, 17. Below we discuss the relevance

RTEL1 and cancer predisposition

Genome-wide association studies (GWAS) established an association of single-nucleotide polymorphisms (SNPs) in RTEL1 with increased susceptibility to brain tumours. A principal component-adjusted GWAS study, comprising over 275 000 autosomal variants among 692 adult glioma cases and 3992 controls, identified two SNPs within intron 12 (rs6010620) and intron 17 (rs4809324) of RTEL1 that are significantly associated with glioma and astrocytoma predisposition [47]. Similarly, two further glioma GWAS

RTEL1 and Hoyeraal–Hreidarsson syndrome

Telomere attrition, a natural biological process that arises from failure to maintain telomere homeostasis, has been linked to an elevated risk of a variety of age-related diseases (reviewed in 63, 64, 65, 66). Critically short telomeres signal the cell to terminate division and enter a stable cell cycle arrest termed senescence (reviewed in 67, 68). Mutations in genes encoding telomere-associated proteins give rise to premature aging disorders such as Werner Syndrome, Hoyeraal–Hreidarsson

Concluding remarks

Although recent insights into the function of RTEL1 in controlling HR, telomere homeostasis and facilitating DNA replication have highlighted its importance in the maintenance of genome stability, many outstanding questions remain to be addressed. Currently, very little is known about how RTEL1 is regulated or how it is recruited to replication forks and telomeres to execute its functions. The association of RTEL1 with the replisome is evident from its ability to bind directly to PCNA, but the

References (94)

  • M.L. Bochman

    DNA secondary structures: stability and function of G-quadruplex structures

    Nat. Rev. Genet.

    (2012)
  • E. Kruisselbrink

    Mutagenic capacity of endogenous G4 DNA underlies genome instability in FANCJ-defective C. elegans

    Curr. Biol.

    (2008)
  • K. Paeschke

    DNA replication through G-quadruplex motifs is promoted by the Saccharomyces cerevisiae Pif1 DNA helicase

    Cell

    (2011)
  • J.D. Griffith

    Mammalian telomeres end in a large duplex loop

    Cell

    (1999)
  • R.C. Wang

    Homologous recombination generates T-loop-sized deletions at human telomeres

    Cell

    (2004)
  • T. de Lange

    Shelterin: the protein complex that shapes and safeguards human telomeres

    Genes Dev.

    (2005)
  • T. Sugiyama

    A single-stranded DNA-binding protein is needed for efficient presynaptic complex formation by the Saccharomyces cerevisiae Rad51 protein

    J. Biol. Chem.

    (1997)
  • T. Ogawa

    Similarity of the yeast RAD51 filament to the bacterial RecA filament

    Science

    (1993)
  • P. Sung et al.

    DNA strand exchange mediated by a RAD51–ssDNA nucleoprotein filament with polarity opposite to that of RecA

    Cell

    (1995)
  • J.M. Svendsen

    Mammalian BTBD12/SLX4 assembles a Holliday junction resolvase and is required for DNA repair

    Cell

    (2009)
  • Y.C. Lo

    Sgs1 regulates gene conversion tract lengths and crossovers independently of its helicase activity

    Mol. Cell. Biol.

    (2006)
  • L. Wu

    BLAP75/RMI1 promotes the BLM-dependent dissolution of homologous recombination intermediates

    Proc. Natl. Acad. Sci. U.S.A.

    (2006)
  • J.B. Vannier

    RTEL1 dismantles T loops and counteracts telomeric G4-DNA to maintain telomere integrity

    Cell

    (2012)
  • B.J. Ballew

    A recessive founder mutation in regulator of telomere elongation helicase 1, RTEL1, underlies severe immunodeficiency and features of Hoyeraal–Hreidarsson syndrome

    PLoS Genet.

    (2013)
  • B.J. Ballew

    Germline mutations of regulator of telomere elongation helicase 1, RTEL1, in dyskeratosis congenita

    Hum. Genet.

    (2013)
  • Z. Deng

    Inherited mutations in the helicase RTEL1 cause telomere dysfunction and Hoyeraal–Hreidarsson syndrome

    Proc. Natl. Acad. Sci. U.S.A.

    (2013)
  • T. Le Guen

    Human RTEL1 deficiency causes Hoyeraal–Hreidarsson syndrome with short telomeres and genome instability

    Hum. Mol. Genet.

    (2013)
  • A.J. Walne

    Constitutional mutations in RTEL1 cause severe dyskeratosis congenita

    Am. J. Hum. Genet.

    (2013)
  • M.E. Fairman-Williams

    SF1 and SF2 helicases: family matters

    Curr. Opin. Struct. Biol.

    (2010)
  • A. Marchler-Bauer

    CDD: specific functional annotation with the Conserved Domain Database

    Nucleic Acids Res.

    (2009)
  • M.F. White

    Structure, function and evolution of the XPD family of iron–sulfur-containing 5′–>3′ DNA helicases

    Biochem. Soc. Trans.

    (2009)
  • K. Gari

    MMS19 links cytoplasmic iron–sulfur cluster assembly to DNA metabolism

    Science

    (2012)
  • O. Stehling

    MMS19 assembles iron–sulfur proteins required for DNA metabolism and genomic integrity

    Science

    (2012)
  • L. Zhu

    Telomere length regulation in mice is linked to a novel chromosome locus

    Proc. Natl. Acad. Sci. U.S.A.

    (1998)
  • H. Ding

    Regulation of murine telomere length by Rtel: an essential gene encoding a helicase-like protein

    Cell

    (2004)
  • E.J. Uringa

    RTEL1 contributes to DNA replication and repair and telomere maintenance

    Mol. Biol. Cell

    (2012)
  • J.B. Vannier

    RTEL1 is a replisome-associated helicase that promotes telomere and genome-wide replication

    Science

    (2013)
  • L.J. Barber

    RTEL1 maintains genomic stability by suppressing homologous recombination

    Cell

    (2008)
  • P. Sung et al.

    Mechanism of homologous recombination: mediators and helicases take on regulatory functions

    Nat. Rev. Mol. Cell Biol.

    (2006)
  • D.G. Mets et al.

    Condensins regulate meiotic DNA break distribution, thus crossover frequency, by controlling chromosome structure

    Cell

    (2009)
  • J.L. Youds

    RTEL-1 enforces meiotic crossover interference and homeostasis

    Science

    (2010)
  • S. Rosu

    Robust crossover assurance and regulated interhomolog access maintain meiotic crossover number

    Science

    (2011)
  • Y. Doksani

    Super-resolution fluorescence imaging of telomeres reveals TRF2-dependent T-loop formation

    Cell

    (2013)
  • S. Amiard

    A topological mechanism for TRF2-enhanced strand invasion

    Nat. Struct. Mol. Biol.

    (2007)
  • E. Gilson et al.

    How telomeres are replicated

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • B. Wan

    SLX4 assembles a telomere maintenance toolkit by bridging multiple endonucleases with telomeres

    Cell Rep.

    (2013)
  • J.S. Wilson

    Localization-dependent and -independent roles of SLX4 in regulating telomeres

    Cell Rep.

    (2013)
  • A. Sfeir

    Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication

    Cell

    (2009)
  • S.G. Durkin et al.

    Chromosome fragile sites

    Annu. Rev. Genet.

    (2007)
  • J.O. Andressoo

    Nucleotide excision repair and its connection with cancer and ageing

    Adv. Exp. Med. Biol.

    (2005)
  • W.L. Bridge

    The BRIP1 helicase functions independently of BRCA1 in the Fanconi anemia pathway for DNA crosslink repair

    Nat. Genet.

    (2005)
  • M. Levitus

    The DNA helicase BRIP1 is defective in Fanconi anemia complementation group

    J. Nat. Genet.

    (2005)
  • O. Levran

    The BRCA1-interacting helicase BRIP1 is deficient in Fanconi anemia

    Nat. Genet.

    (2005)
  • R. Litman

    BACH1 is critical for homologous recombination and appears to be the Fanconi anemia gene product FANCJ

    Cancer Cell

    (2005)
  • J.M. Capo-Chichi

    Identification and biochemical characterization of a novel mutation in DDX11 causing Warsaw breakage syndrome

    Hum. Mutat.

    (2013)
  • P. van der Lelij

    Warsaw breakage syndrome, a cohesinopathy associated with mutations in the XPD helicase family member DDX11/ChlR1

    Am. J. Hum. Genet.

    (2010)
  • M. Wrensch

    Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility

    Nat. Genet.

    (2009)
  • Cited by (101)

    • Alternative complexes formed by the Escherichia coli clamp loader accessory protein HolC (x) with replication protein HolD (ψ) and repair protein YoaA

      2021, DNA Repair
      Citation Excerpt :

      Eukaryotes and archaea possess multiple members of Fe-S cluster DNA helicase group [26,61] with similar functions in repair and maintenance of genomic stability. In humans, these pathways include excision repair and transcription-couple repair (XPD) [70], DNA cross-link repair (FANCJ, DDX11) [71,72], disassembly of DNA secondary structure and telomere maintenance (RTEL, FANCJ) [73,74], and homologous recombination (RTEL, DDX11) [72,74]. Defects in these underlie a number of genetic diseases, including XPD xeroderma pigmentosum, Cockayne syndrome and trichothiodystrophy; FANC-J, Fanconi anemia and breast cancer proneness; RTEL, Hoyeraal-Hreidarsson syndrome; DDX11, Warsaw breakage syndrome.

    • Molecular mechanisms of telomere biology disorders

      2021, Journal of Biological Chemistry
    • RTEL1 Regulates G4/R-Loops to Avert Replication-Transcription Collisions

      2020, Cell Reports
      Citation Excerpt :

      Subsequent genome-wide association studies identified RTEL1 as a susceptibility locus for astrocytomas, high-grade gliomas, and many other cancers. Hypomorphic mutations in human RTEL1 are also causal for Hoyeraal-Hreidarsson syndrome (HHS), a severe disorder associated with inter-uterine growth retardation, microcephaly, bone marrow failure, immunodeficiency, and many other complications (for review, see Vannier et al., 2014). While the etiology of HHS remains to be fully elucidated, patient-derived cells and PIP box knockin mouse cells present with both telomeric attrition, increased replication stress, and reduced proliferative capacity in culture.

    View all citing articles on Scopus
    *

    These authors contributed equally.

    View full text