SlideShare a Scribd company logo
AUTHOR’S VIEW
Retinoblastoma family proteins: New players in DNA repair by non-homologous
end-joining
Paul H. Huanga
, Rebecca Cooka,b
, Georgia Zoumpoulidoua
, Maciej T. Luczynskia
, and Sibylle Mittnachta,b
a
The Institute of Cancer Research, Division of Cancer Biology, Chester Beatty Laboratories, London, UK; b
Cancer Cell Signalling, UCL Cancer Institute,
University College London, London, UK
ARTICLE HISTORY
Received 15 May 2015
Revised 17 May 2015
Accepted 18 May 2015
ABSTRACT
Loss of retinoblastoma protein (RB1) function is a major driver in cancer development. We have recently
reported that, in addition to its well-documented functions in cell cycle and fate control, RB1 and its
paralogs have a novel role in regulating DNA repair by non-homologous end joining (NHEJ). Here we
summarize our findings and present mechanistic hypotheses on how RB1 may support the DNA repair
process and the therapeutic implications for patients who harbor RB1-negative cancers.
KEYWORDS
DNA repair; non-homologous
end-joining; retinoblastoma
protein; tumor suppressor
The tumor suppressor retinoblastoma protein (RB1) is a critical
component of proliferation and fate control in cells.1,2
Heritable
mutations in RB1 cause a highly penetrant predisposition to the
childhood cancer retinoblastoma, combined with a substan-
tially increased lifetime risk for other cancers.3
Sporadic muta-
tions are associated with a spectrum of difficult-to-treat cancers
including bone and soft tissue sarcoma, small cell lung cancer,
breast, endometrial, ovarian, and renal carcinoma.
RB1’s functions rely on its interaction with cellular pro-
teins,4
with most known proteins associating with the central
A-B pocket (RB1P
; amino acid [aa] 379-792) and the RB1C
domain (aa 792-928). These include proteins containing a Leu-
X-Cys-X-Glu (LXCXE) short linear motif and members of the
E2F family of transcription factors, through which RB1 sup-
presses gene transcription and effects cell cycle control, mitotic
fidelity, and cell fate responses (Fig. 1).5-10
Despite evidence for
involvement of the third functional domain, RB1N
(aa 1-379),
in RB1 protein function and tumor suppression,11
the molecu-
lar basis through which this domain contributes to these activi-
ties has hitherto remained undiscovered. Although the
majority of RB1 mutations result in radical protein loss, in both
childhood retinoblastoma (http://rb1-lsdb.d-lohmann.de/) and
major cancers such as small cell lung cancer and bladder cancer
(www.cbioportal.com) some mutations, including small altera-
tions within the RB1N
domain, are frame preserving.
Previous work from our laboratory reporting the crystal
structure of RB1N
revealed a striking similarity between RB1N
and RB1P
, predicting an evolutionary relationship and concep-
tual similarity in function between these 2 RB1 domains.12
Like
RB1P
, RB1N
contains 2 tandem cyclin-like folds, and significant
amino acid identity within the corresponding cyclin fold helices
suggests that RB1 may have evolved through duplication of an
ancestral cyclin fold pair (Fig. 1). Importantly, a surface iso-
structural to the ‘cyclin wedge’ through which the cellular
cyclins interact with their partner kinase subunits, and which
in RB1P
is used for the binding of LXCXE proteins,7
is present
in RB1N
and overt residue conservation within this surface sug-
gests that it is involved in the interaction of RB1N
with cellular
partner proteins.
Given such structural evidence, together with longstanding
documentation that RB1N
has the capacity to bind partner pro-
teins in vitro and carries mutations in tumors, we hypothesized
that RB1N
may mediate tumor suppressor functions through
interactions with cellular partner proteins in a similar manner
to RB1P
. Building on this notion, in our recently published
study13
we probed cellular nuclear extracts using affinity purifi-
cation mass spectrometry. In this proteomic screen, 34 proteins
across a range of biologic functions were found to selectively
interact with RB1N
.13
Among them were several proteins func-
tionally involved in double-strand break DNA repair by non-
homologous end joining (NHEJ), namely X-ray repair comple-
menting defective repair in Chinese hamster cells 5 and 6
(XRCC5 and XRCC6), which are required for the recognition
of DNA double strand breaks (DSBs), and the double-strand
DNA-dependent protein kinase PRKDC (protein kinase,
DNA-activated, catalytic polypeptide).14
Structure-guided
mutations demonstrated that the cyclin wedge homology sur-
face within RB1N
is required for binding of XRCC5 and
XRCC6, indicating that this specific RB1N
surface, which is pre-
dicted to facilitate protein interactions, is responsible for medi-
ating the interaction with NHEJ components.
NHEJ is a prominent pathway in DSB DNA repair and facil-
itates the rapid joining of broken DNA ends without the need
for a homologous repair template.15
NHEJ is critical for the
faithful repair of DNA damage during GAP1 (G1) and early S
phase of the cell cycle and for the repair of complex DNA dam-
age throughout the cell cycle. Our further functional assess-
ment provides support for the direct functional involvement of
CONTACT Sibylle Mittnacht s.mittnacht@ucl.ac.uk; Paul H. Huang paul.huang@icr.ac.uk
© 2016 Taylor & Francis Group, LLC
MOLECULAR & CELLULAR ONCOLOGY
2016, VOL. 3, NO. 2, e1053596 (3 pages)
http://dx.doi.org/10.1080/23723556.2015.1053596
RB1 in DNA repair by NHEJ with additional evidence that it
shares this function with its paralogs p107 and p130. Notably, 2
RB1 variants that possessed mutations in the cyclin wedge
homology region and were therefore unable to physically inter-
act with XRCC6 could not reinstate NHEJ when expressed in
RB1-negative cells. Neither of these RB1 variants had a detect-
able defect in inhibition of cell cycle progression, reduction of
colony numbers, or suppression of transcription via E2F, indi-
cating that the roles of RB1 in proliferation control and DNA
repair are genetically and functionally separable and indepen-
dent. We further demonstrate that RB loss adversely affects
chromosomal integrity upon irradiation. Specifically, RB loss
increased the frequency of chromosomal aberrations in cells
independent of mitotic transit, a recognized consequence of
defective NHEJ. Together, our data argue that RB1 and its
paralogs act to enable NHEJ through physical interaction with
components of the canonical NHEJ pathway, and that this
activity is unrelated to the canonical cell cycle and transcription
regulatory functions of RB1 family proteins.
Outstanding questions remain regarding the mechanism(s)
by which the RB1 family proteins support the DNA repair pro-
cess. It is thought that RB1 orchestrates the assembly of
multiprotein complexes in cells through coordinate use of pro-
tein binding surfaces. Both RB1P
and RB1C
interact with chro-
matin modifiers recognized for their contribution in gene
regulation and the regulation of chromatin conformation.10,16
Many of these same modifiers are also known to be involved in
NHEJ. These include histone deacetylase 1 and 2 (HDAC 1 and
2) and the histone methyltransferase SUV4 (suppressor of var-
iegation 4-20 homolog 1), which lead to chromatin condensa-
tion.17-19
A concurrently published report documents
interaction of RB1C
with the tudor domain of tumor protein 53
binding protein 1 (TP53BP1),20
which is involved in suppress-
ing the 50
resection of broken DNA ends, in turn discouraging
the use of homology driven repair events (Fig. 1B). Thus, a
tempting mechanistic hypothesis would be that RB1N
, RB1P
,
and RB1C
enable NHEJ DNA repair in a cooperative manner.
In the first stage, RB1 may interact with complexes containing
XRCC6 via the RB1N
domain, which then may serves as a plat-
form for the subsequent recruitment of chromatin modifiers to
the sites of damage by RB1P
and RB1C
.
Since RB1 loss leads to defects in canonical NHEJ, RB1-neg-
ative cancers are likely to be more reliant on other forms of
DNA double-strand break repair, including homologous
Figure 1. Domains and binding partners of retinoblastoma protein (RB1). (A) Domain organization of RB1, a 928-amino acid protein composed of 3 distinct folded
domains, and (B) structural model of RB1 depicting the individual folded domains with annotation of positions of protein binding. Cyclin-like fold pair of the N-terminal
domain, RB1N
(2QDJ) is colored green, cyclin-like fold pair of the central pocket domain RB1P
(3POM) is colored purple, and the C-terminal domain RB1C
(2AZE) is colored
yellow. The relative positioning of RBN
and RBP
is described in ref. 24.24
The relative positioning of RBC
is inferred by the positioning of the RBP
COOH-terminus. Proteins
known to bind to distinct regions of RB1 are highlighted in blue boxes. RB1 plays a functional role in transcriptional control of cell proliferation through binding to E2F
and Leu-X-Cys-X-Glu (LXCXE) proteins. In addition, RB1 regulates DNA damage repair through the binding of non-homologous end-joining (NHEJ) proteins in the N-termi-
nal domain. By simultaneously binding multiple proteins, such as histone deacetylases (HDACs) via the central pocket domain and the DNA damage repair protein tumor
protein p65 binding protein 1 (TP53BP1) via methylated K810 (K810Me) in the C-terminal domain, RB1 may facilitate the recruitment of chromatin regulatory proteins to
sites of DNA damage, in turn creating a chromatin environment supportive of DNA damage repair by NHEJ. CTBP, C-terminal binding protein; DP1, DP1 transcription fac-
tor; E2F, E2F transcription factor; SIN3A, SIN3 transcriptional regulator family member A; XRCC5/6, X-ray repair complementing defective repair in Chinese hamster cells 5
and 6.
e1053596-2 P. H. HUANG ET AL.
recombination (HR) and alternative inaccurate forms of NHEJ
(aNHEJ).21,22
We can thus frame a clinical hypothesis whereby
dependencies on alternative repair may identify therapeutic
strategies that specifically sensitize cancers with RB1 loss to
genotoxic stress. Our findings would predict that inhibition of
such repair routes in RB1-defective cancers would offer a thera-
peutic opportunity by promoting hypersensitivity in these can-
cers to therapeutic regimens of DNA damage. Loss of RB1
protein function is found in a number of cancers with few
options for effective treatment, including small cell lung cancer,
ovarian cancer, sarcoma, and triple negative breast cancer.
Analogous to the success of poly (ADP-ribose) polymerase
(PARP) inhibitors in BRCA1 and BRCA2 (breast cancer 1 and
2, early onset)-deficient patients that display defects in HR-
mediated DNA repair,23
future work to characterize the molec-
ular mechanisms of DNA repair in RB1-defective cancers may
facilitate the identification of novel therapeutic approaches for
patients harboring RB1-negative tumors.
Disclosure of potential conflicts of interest
No potential conflicts of interest were disclosed.
Funding
The work was supported through Cancer Research UK (grant C107/10433
and CRUK/A15043 to S.M.) and Worldwide Cancer Research (grant num-
ber 12-1280 to S.M.). R.C. was supported by an ICR student fellowship. P.
H.H. is supported by the Wellcome Trust (WT089028).
References
1. Classon M, Harlow E. The retinoblastoma tumour suppressor in
development and cancer. Nat Rev Cancer 2002; 2:910-7;
PMID:12459729; http://dx.doi.org/10.1038/nrc950
2. Khidr L, Chen PL. RB, the conductor that orchestrates life, death and
differentiation. Oncogene 2006; 25:5210-9; PMID:16936739; http://
dx.doi.org/10.1038/sj.onc.1209612
3. Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert
DM, Dryja TP. A human DNA segment with properties of the gene
that predisposes to retinoblastoma and osteosarcoma. Nature 1986;
323:643-6; PMID:2877398; http://dx.doi.org/10.1038/323643a0
4. Morris EJ, Dyson NJ. Retinoblastoma protein partners. Adv Cancer
Res 2001; 82:1-54; PMID:11447760
5. Ferreira R, Magnaghi-Jaulin L, Robin P, Harel-Bellan A, Trouche D.
The three members of the pocket proteins family share the ability to
repress E2F activity through recruitment of a histone deacetylase.
Proc Natl Acad Sci U S A 1998; 95:10493-8; PMID:9724731; http://dx.
doi.org/10.1073/pnas.95.18.10493
6. Rubin SM, Gall AL, Zheng N, Pavletich NP. Structure of the Rb C-ter-
minal domain bound to E2F1-DP1: a mechanism for phosphoryla-
tion-induced E2F release. Cell 2005; 123:1093-106; PMID:16360038;
http://dx.doi.org/10.1016/j.cell.2005.09.044
7. Lee JO, Russo AA, Pavletich NP. Structure of the retinoblastoma tumour-
suppressor pocket domain bound to a peptide from HPV E7. Nature 1998;
391:859-65; PMID:9495340; http://dx.doi.org/10.1038/36038
8. Lee C, Chang JH, Lee HS, Cho Y. Structural basis for the recognition
of the E2F transactivation domain by the retinoblastoma tumor sup-
pressor. Genes Dev 2002; 16:3199-212; PMID:12502741; http://dx.doi.
org/10.1101/gad.1046102
9. Xiao B, Spencer J, Clements A, Ali-Khan N, Mittnacht S, Broceno C,
Burghammer M, Perrakis A, Marmorstein R, Gamblin SJ. Crystal
structure of the retinoblastoma tumor suppressor protein bound to
E2F and the molecular basis of its regulation. Proc Natl Acad Sci U S
A 2003; 100:2363-8; PMID:12598654; http://dx.doi.org/10.1073/
pnas.0436813100
10. Talluri S, Dick FA. Regulation of transcription and chromatin struc-
ture by pRB: here, there and everywhere. Cell Cycle 2012; 11:3189-98;
PMID:22895179; http://dx.doi.org/10.4161/cc.21263
11. Goodrich DW. How the other half lives, the amino-terminal
domain of the retinoblastoma tumor suppressor protein. J Cell
Physiol 2003; 197:169-80; PMID:14502556; http://dx.doi.org/
10.1002/jcp.10358
12. Hassler M, Singh S, Yue WW, Luczynski M, Lakbir R, Sanchez-San-
chez F, Bader T, Pearl LH, Mittnacht S. Crystal structure of the retino-
blastoma protein N domain provides insight into tumor suppression,
ligand interaction, and holoprotein architecture. Mol Cell 2007;
28:371-85; PMID:17996702; http://dx.doi.org/10.1016/j.
molcel.2007.08.023
13. Cook R, Zoumpoulidou G, Luczynski MT, Rieger S, Moquet J, Spans-
wick VJ, Hartley JA, Rothkamm K, Huang PH, Mittnacht S. Direct
Involvement of Retinoblastoma Family Proteins in DNA Repair by
Non-homologous End-Joining. Cell Rep 2015; 10:2006-18;
PMID:25818292; http://dx.doi.org/10.1016/j.celrep.2015.02.059
14. Lieber MR, Ma Y, Pannicke U, Schwarz K. Mechanism and regulation
of human non-homologous DNA end-joining. Nat Rev Mol Cell Biol
2003; 4:712-20; PMID:14506474; http://dx.doi.org/10.1038/nrm1202
15. Lieber MR. The mechanism of double-strand DNA break repair by the
nonhomologous DNA end-joining pathway. Annu Rev Biochem 2010;
79:181-211; PMID:20192759; http://dx.doi.org/10.1146/annurev.
biochem.052308.093131
16. Manning AL, Dyson NJ. RB: mitotic implications of a tumour sup-
pressor. Nat Rev Cancer 2012; 12:220-6; PMID:22318235
17. Lai A, Lee JM, Yang WM, DeCaprio JA, Kaelin WG, Jr., Seto E, Bran-
ton PE. RBP1 recruits both histone deacetylase-dependent and -inde-
pendent repression activities to retinoblastoma family proteins. Mol
Cell Biol 1999; 19:6632-41; PMID:10490602
18. Gonzalo S, Garcia-Cao M, Fraga MF, Schotta G, Peters AH, Cotter SE,
Eguıa R, Dean DC, Esteller M, Jenuwein T, et al. Role of the RB1 fam-
ily in stabilizing histone methylation at constitutive heterochromatin.
Nat Cell Biol 2005; 7:420-8; PMID:15750587; http://dx.doi.org/
10.1038/ncb1235
19. Isaac CE, Francis SM, Martens AL, Julian LM, Seifried LA, Erdmann
N, Binne UK, Harrington L, Sicinski P, Berube NG, et al. The retino-
blastoma protein regulates pericentric heterochromatin. Mol Cell Biol
2006; 26:3659-71; PMID:16612004; http://dx.doi.org/10.1128/
MCB.26.9.3659-3671.2006
20. Carr SM, Munro S, Zalmas LP, Fedorov O, Johansson C, Krojer T,
Sagum CA, Bedford MT, Oppermann U, La Thangue NB. Lysine
methylation-dependent binding of 53BP1 to the pRb tumor suppres-
sor. Proc Natl Acad Sci U S A 2014; 111:11341-6; PMID:25049398;
http://dx.doi.org/10.1073/pnas.1403737111
21. Aparicio T, Baer R, Gautier J. DNA double-strand break repair path-
way choice and cancer. DNA Repair (Amst) 2014; 19:169-75;
PMID:24746645; http://dx.doi.org/10.1016/j.dnarep.2014.03.014
22. Heyer WD, Ehmsen KT, Liu J. Regulation of homologous recombina-
tion in eukaryotes. Annu Rev Genet 2010; 44:113-39;
PMID:20690856; http://dx.doi.org/10.1146/annurev-genet-051710-
150955
23. Lord CJ, Ashworth A. Targeted therapy for cancer using PARP inhibi-
tors. Curr Opin Pharmacol 2008; 8:363-9; PMID:18644251; http://dx.
doi.org/10.1016/j.coph.2008.06.016
24. Lamber EP, Beuron F, Morris EP, Svergun DI, Mittnacht S. Structural
insights into the mechanism of phosphoregulation of the retinoblas-
toma protein. PLoS One 2013; 8:e58463; PMID:23516486; http://dx.
doi.org/10.1371/journal.pone.0058463
MOLECULAR  CELLULAR ONCOLOGY e1053596-3

More Related Content

What's hot

Structural Mechanism for the Fidelity Modulation of DNA Polymerase λ
Structural Mechanism for the Fidelity Modulation of DNA Polymerase λStructural Mechanism for the Fidelity Modulation of DNA Polymerase λ
Structural Mechanism for the Fidelity Modulation of DNA Polymerase λ
Mu-Sen Liu
 
Presentazione I anno Federica Campana
Presentazione I anno Federica CampanaPresentazione I anno Federica Campana
Presentazione I anno Federica Campana
lab13unisa
 
Analysis of metabolic pathways in relation to the
Analysis of metabolic pathways in relation to theAnalysis of metabolic pathways in relation to the
Analysis of metabolic pathways in relation to the
Aishwarya Raj
 
Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...
Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...
Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...
Denise Sheer
 
Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009
Roberto Perales PhD
 
Voss et al. - 2006 - Identification and characterization of riproximin,
Voss et al. - 2006 - Identification and characterization of riproximin,Voss et al. - 2006 - Identification and characterization of riproximin,
Voss et al. - 2006 - Identification and characterization of riproximin,
Cristina Voss
 

What's hot (20)

biomedicina_Dolzan_TransporterVariability.pdf
biomedicina_Dolzan_TransporterVariability.pdfbiomedicina_Dolzan_TransporterVariability.pdf
biomedicina_Dolzan_TransporterVariability.pdf
 
CBO9780511781933.008.pdf
CBO9780511781933.008.pdfCBO9780511781933.008.pdf
CBO9780511781933.008.pdf
 
SOAR Poster Final
SOAR Poster FinalSOAR Poster Final
SOAR Poster Final
 
Wang_NSMB2009
Wang_NSMB2009Wang_NSMB2009
Wang_NSMB2009
 
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
Molecular biology revision-Part 3 (Regulation of genes expression and Recombi...
 
Sigma Xi Presentation
Sigma Xi PresentationSigma Xi Presentation
Sigma Xi Presentation
 
Structural Mechanism for the Fidelity Modulation of DNA Polymerase λ
Structural Mechanism for the Fidelity Modulation of DNA Polymerase λStructural Mechanism for the Fidelity Modulation of DNA Polymerase λ
Structural Mechanism for the Fidelity Modulation of DNA Polymerase λ
 
CYTOKINES RECEPTOR
CYTOKINES RECEPTORCYTOKINES RECEPTOR
CYTOKINES RECEPTOR
 
Presentazione I anno Federica Campana
Presentazione I anno Federica CampanaPresentazione I anno Federica Campana
Presentazione I anno Federica Campana
 
Analysis of metabolic pathways in relation to the
Analysis of metabolic pathways in relation to theAnalysis of metabolic pathways in relation to the
Analysis of metabolic pathways in relation to the
 
Lab5_NguyenAlbert
Lab5_NguyenAlbertLab5_NguyenAlbert
Lab5_NguyenAlbert
 
Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...
Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...
Microhomology in Genomic Stuctural Variation - Diego Ottaviani, Magdalena Lec...
 
Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009Perales and Bentley MolCell 2009
Perales and Bentley MolCell 2009
 
Voss et al. - 2006 - Identification and characterization of riproximin,
Voss et al. - 2006 - Identification and characterization of riproximin,Voss et al. - 2006 - Identification and characterization of riproximin,
Voss et al. - 2006 - Identification and characterization of riproximin,
 
Histone modifications
Histone modificationsHistone modifications
Histone modifications
 
DNA damage and repair systems
DNA damage and repair systemsDNA damage and repair systems
DNA damage and repair systems
 
Tgf β signalling from cell
Tgf β signalling from cellTgf β signalling from cell
Tgf β signalling from cell
 
finalposterapril13
finalposterapril13finalposterapril13
finalposterapril13
 
Atp binding cassette (abc) transporter
Atp binding cassette (abc) transporterAtp binding cassette (abc) transporter
Atp binding cassette (abc) transporter
 
Seminario biologia molecular. ana maria agudelo
Seminario biologia molecular. ana maria agudeloSeminario biologia molecular. ana maria agudelo
Seminario biologia molecular. ana maria agudelo
 

Similar to Retinoblastoma family proteins New players in DNA repair by non-homologous

Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulator
rkkoiri
 
Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulator
rkkoiri
 
ZechSG16-acs-jmedchem-5b01552
ZechSG16-acs-jmedchem-5b01552ZechSG16-acs-jmedchem-5b01552
ZechSG16-acs-jmedchem-5b01552
Stephan G. Zech
 
Crystal Structure of the Retinoblastoma Protein
Crystal Structure of the Retinoblastoma ProteinCrystal Structure of the Retinoblastoma Protein
Crystal Structure of the Retinoblastoma Protein
Maciej Luczynski
 
Siemens Research Paper FINAL
Siemens Research Paper FINALSiemens Research Paper FINAL
Siemens Research Paper FINAL
Zuhair Mohammed
 
transciption powerpoint
transciption powerpointtransciption powerpoint
transciption powerpoint
Nikka Bañez
 
Horrix et al. - 2010
Horrix et al. - 2010Horrix et al. - 2010
Horrix et al. - 2010
Cristina Voss
 
PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)
Marco Garza
 
Undergraduate Research Symposium Poster
Undergraduate Research Symposium PosterUndergraduate Research Symposium Poster
Undergraduate Research Symposium Poster
Tim Krueger
 
Biochim Biophys Acta. 2014
Biochim Biophys Acta. 2014Biochim Biophys Acta. 2014
Biochim Biophys Acta. 2014
Laia Llovera
 

Similar to Retinoblastoma family proteins New players in DNA repair by non-homologous (20)

Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulator
 
Ruthenium complex as enzyme modulator
Ruthenium complex as enzyme modulatorRuthenium complex as enzyme modulator
Ruthenium complex as enzyme modulator
 
Single Nucleotide Polymorphism - The new generation therapy
Single Nucleotide Polymorphism - The new generation therapySingle Nucleotide Polymorphism - The new generation therapy
Single Nucleotide Polymorphism - The new generation therapy
 
ZechSG16-acs-jmedchem-5b01552
ZechSG16-acs-jmedchem-5b01552ZechSG16-acs-jmedchem-5b01552
ZechSG16-acs-jmedchem-5b01552
 
71st ICREA Colloquium "Intrinsically disordered proteins (id ps) the challeng...
71st ICREA Colloquium "Intrinsically disordered proteins (id ps) the challeng...71st ICREA Colloquium "Intrinsically disordered proteins (id ps) the challeng...
71st ICREA Colloquium "Intrinsically disordered proteins (id ps) the challeng...
 
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
71st ICREA Colloquium - Intrinsically disordered proteins (IDPs) the challeng...
 
16. Biology of Cancer
16. Biology of Cancer16. Biology of Cancer
16. Biology of Cancer
 
Crystal Structure of the Retinoblastoma Protein
Crystal Structure of the Retinoblastoma ProteinCrystal Structure of the Retinoblastoma Protein
Crystal Structure of the Retinoblastoma Protein
 
Molecular cell biology 2 transcription
Molecular cell biology 2 transcriptionMolecular cell biology 2 transcription
Molecular cell biology 2 transcription
 
Siemens Research Paper FINAL
Siemens Research Paper FINALSiemens Research Paper FINAL
Siemens Research Paper FINAL
 
transciption powerpoint
transciption powerpointtransciption powerpoint
transciption powerpoint
 
Identification and Characterization of Saccharomyces cerevisiae Cdc6 DNA-bind...
Identification and Characterization of Saccharomyces cerevisiae Cdc6 DNA-bind...Identification and Characterization of Saccharomyces cerevisiae Cdc6 DNA-bind...
Identification and Characterization of Saccharomyces cerevisiae Cdc6 DNA-bind...
 
Horrix et al. - 2010
Horrix et al. - 2010Horrix et al. - 2010
Horrix et al. - 2010
 
Seah_SURF (1)
Seah_SURF (1)Seah_SURF (1)
Seah_SURF (1)
 
PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)PIIS1552526009012771(1)(3)
PIIS1552526009012771(1)(3)
 
LSD1 - bmc-paper
LSD1 - bmc-paperLSD1 - bmc-paper
LSD1 - bmc-paper
 
nihms336576
nihms336576nihms336576
nihms336576
 
Undergraduate Research Symposium Poster
Undergraduate Research Symposium PosterUndergraduate Research Symposium Poster
Undergraduate Research Symposium Poster
 
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
CELLULAR REPROGRAMMING: Current Technology, Perspectives and Generation of iP...
 
Biochim Biophys Acta. 2014
Biochim Biophys Acta. 2014Biochim Biophys Acta. 2014
Biochim Biophys Acta. 2014
 

Retinoblastoma family proteins New players in DNA repair by non-homologous

  • 1. AUTHOR’S VIEW Retinoblastoma family proteins: New players in DNA repair by non-homologous end-joining Paul H. Huanga , Rebecca Cooka,b , Georgia Zoumpoulidoua , Maciej T. Luczynskia , and Sibylle Mittnachta,b a The Institute of Cancer Research, Division of Cancer Biology, Chester Beatty Laboratories, London, UK; b Cancer Cell Signalling, UCL Cancer Institute, University College London, London, UK ARTICLE HISTORY Received 15 May 2015 Revised 17 May 2015 Accepted 18 May 2015 ABSTRACT Loss of retinoblastoma protein (RB1) function is a major driver in cancer development. We have recently reported that, in addition to its well-documented functions in cell cycle and fate control, RB1 and its paralogs have a novel role in regulating DNA repair by non-homologous end joining (NHEJ). Here we summarize our findings and present mechanistic hypotheses on how RB1 may support the DNA repair process and the therapeutic implications for patients who harbor RB1-negative cancers. KEYWORDS DNA repair; non-homologous end-joining; retinoblastoma protein; tumor suppressor The tumor suppressor retinoblastoma protein (RB1) is a critical component of proliferation and fate control in cells.1,2 Heritable mutations in RB1 cause a highly penetrant predisposition to the childhood cancer retinoblastoma, combined with a substan- tially increased lifetime risk for other cancers.3 Sporadic muta- tions are associated with a spectrum of difficult-to-treat cancers including bone and soft tissue sarcoma, small cell lung cancer, breast, endometrial, ovarian, and renal carcinoma. RB1’s functions rely on its interaction with cellular pro- teins,4 with most known proteins associating with the central A-B pocket (RB1P ; amino acid [aa] 379-792) and the RB1C domain (aa 792-928). These include proteins containing a Leu- X-Cys-X-Glu (LXCXE) short linear motif and members of the E2F family of transcription factors, through which RB1 sup- presses gene transcription and effects cell cycle control, mitotic fidelity, and cell fate responses (Fig. 1).5-10 Despite evidence for involvement of the third functional domain, RB1N (aa 1-379), in RB1 protein function and tumor suppression,11 the molecu- lar basis through which this domain contributes to these activi- ties has hitherto remained undiscovered. Although the majority of RB1 mutations result in radical protein loss, in both childhood retinoblastoma (http://rb1-lsdb.d-lohmann.de/) and major cancers such as small cell lung cancer and bladder cancer (www.cbioportal.com) some mutations, including small altera- tions within the RB1N domain, are frame preserving. Previous work from our laboratory reporting the crystal structure of RB1N revealed a striking similarity between RB1N and RB1P , predicting an evolutionary relationship and concep- tual similarity in function between these 2 RB1 domains.12 Like RB1P , RB1N contains 2 tandem cyclin-like folds, and significant amino acid identity within the corresponding cyclin fold helices suggests that RB1 may have evolved through duplication of an ancestral cyclin fold pair (Fig. 1). Importantly, a surface iso- structural to the ‘cyclin wedge’ through which the cellular cyclins interact with their partner kinase subunits, and which in RB1P is used for the binding of LXCXE proteins,7 is present in RB1N and overt residue conservation within this surface sug- gests that it is involved in the interaction of RB1N with cellular partner proteins. Given such structural evidence, together with longstanding documentation that RB1N has the capacity to bind partner pro- teins in vitro and carries mutations in tumors, we hypothesized that RB1N may mediate tumor suppressor functions through interactions with cellular partner proteins in a similar manner to RB1P . Building on this notion, in our recently published study13 we probed cellular nuclear extracts using affinity purifi- cation mass spectrometry. In this proteomic screen, 34 proteins across a range of biologic functions were found to selectively interact with RB1N .13 Among them were several proteins func- tionally involved in double-strand break DNA repair by non- homologous end joining (NHEJ), namely X-ray repair comple- menting defective repair in Chinese hamster cells 5 and 6 (XRCC5 and XRCC6), which are required for the recognition of DNA double strand breaks (DSBs), and the double-strand DNA-dependent protein kinase PRKDC (protein kinase, DNA-activated, catalytic polypeptide).14 Structure-guided mutations demonstrated that the cyclin wedge homology sur- face within RB1N is required for binding of XRCC5 and XRCC6, indicating that this specific RB1N surface, which is pre- dicted to facilitate protein interactions, is responsible for medi- ating the interaction with NHEJ components. NHEJ is a prominent pathway in DSB DNA repair and facil- itates the rapid joining of broken DNA ends without the need for a homologous repair template.15 NHEJ is critical for the faithful repair of DNA damage during GAP1 (G1) and early S phase of the cell cycle and for the repair of complex DNA dam- age throughout the cell cycle. Our further functional assess- ment provides support for the direct functional involvement of CONTACT Sibylle Mittnacht s.mittnacht@ucl.ac.uk; Paul H. Huang paul.huang@icr.ac.uk © 2016 Taylor & Francis Group, LLC MOLECULAR & CELLULAR ONCOLOGY 2016, VOL. 3, NO. 2, e1053596 (3 pages) http://dx.doi.org/10.1080/23723556.2015.1053596
  • 2. RB1 in DNA repair by NHEJ with additional evidence that it shares this function with its paralogs p107 and p130. Notably, 2 RB1 variants that possessed mutations in the cyclin wedge homology region and were therefore unable to physically inter- act with XRCC6 could not reinstate NHEJ when expressed in RB1-negative cells. Neither of these RB1 variants had a detect- able defect in inhibition of cell cycle progression, reduction of colony numbers, or suppression of transcription via E2F, indi- cating that the roles of RB1 in proliferation control and DNA repair are genetically and functionally separable and indepen- dent. We further demonstrate that RB loss adversely affects chromosomal integrity upon irradiation. Specifically, RB loss increased the frequency of chromosomal aberrations in cells independent of mitotic transit, a recognized consequence of defective NHEJ. Together, our data argue that RB1 and its paralogs act to enable NHEJ through physical interaction with components of the canonical NHEJ pathway, and that this activity is unrelated to the canonical cell cycle and transcription regulatory functions of RB1 family proteins. Outstanding questions remain regarding the mechanism(s) by which the RB1 family proteins support the DNA repair pro- cess. It is thought that RB1 orchestrates the assembly of multiprotein complexes in cells through coordinate use of pro- tein binding surfaces. Both RB1P and RB1C interact with chro- matin modifiers recognized for their contribution in gene regulation and the regulation of chromatin conformation.10,16 Many of these same modifiers are also known to be involved in NHEJ. These include histone deacetylase 1 and 2 (HDAC 1 and 2) and the histone methyltransferase SUV4 (suppressor of var- iegation 4-20 homolog 1), which lead to chromatin condensa- tion.17-19 A concurrently published report documents interaction of RB1C with the tudor domain of tumor protein 53 binding protein 1 (TP53BP1),20 which is involved in suppress- ing the 50 resection of broken DNA ends, in turn discouraging the use of homology driven repair events (Fig. 1B). Thus, a tempting mechanistic hypothesis would be that RB1N , RB1P , and RB1C enable NHEJ DNA repair in a cooperative manner. In the first stage, RB1 may interact with complexes containing XRCC6 via the RB1N domain, which then may serves as a plat- form for the subsequent recruitment of chromatin modifiers to the sites of damage by RB1P and RB1C . Since RB1 loss leads to defects in canonical NHEJ, RB1-neg- ative cancers are likely to be more reliant on other forms of DNA double-strand break repair, including homologous Figure 1. Domains and binding partners of retinoblastoma protein (RB1). (A) Domain organization of RB1, a 928-amino acid protein composed of 3 distinct folded domains, and (B) structural model of RB1 depicting the individual folded domains with annotation of positions of protein binding. Cyclin-like fold pair of the N-terminal domain, RB1N (2QDJ) is colored green, cyclin-like fold pair of the central pocket domain RB1P (3POM) is colored purple, and the C-terminal domain RB1C (2AZE) is colored yellow. The relative positioning of RBN and RBP is described in ref. 24.24 The relative positioning of RBC is inferred by the positioning of the RBP COOH-terminus. Proteins known to bind to distinct regions of RB1 are highlighted in blue boxes. RB1 plays a functional role in transcriptional control of cell proliferation through binding to E2F and Leu-X-Cys-X-Glu (LXCXE) proteins. In addition, RB1 regulates DNA damage repair through the binding of non-homologous end-joining (NHEJ) proteins in the N-termi- nal domain. By simultaneously binding multiple proteins, such as histone deacetylases (HDACs) via the central pocket domain and the DNA damage repair protein tumor protein p65 binding protein 1 (TP53BP1) via methylated K810 (K810Me) in the C-terminal domain, RB1 may facilitate the recruitment of chromatin regulatory proteins to sites of DNA damage, in turn creating a chromatin environment supportive of DNA damage repair by NHEJ. CTBP, C-terminal binding protein; DP1, DP1 transcription fac- tor; E2F, E2F transcription factor; SIN3A, SIN3 transcriptional regulator family member A; XRCC5/6, X-ray repair complementing defective repair in Chinese hamster cells 5 and 6. e1053596-2 P. H. HUANG ET AL.
  • 3. recombination (HR) and alternative inaccurate forms of NHEJ (aNHEJ).21,22 We can thus frame a clinical hypothesis whereby dependencies on alternative repair may identify therapeutic strategies that specifically sensitize cancers with RB1 loss to genotoxic stress. Our findings would predict that inhibition of such repair routes in RB1-defective cancers would offer a thera- peutic opportunity by promoting hypersensitivity in these can- cers to therapeutic regimens of DNA damage. Loss of RB1 protein function is found in a number of cancers with few options for effective treatment, including small cell lung cancer, ovarian cancer, sarcoma, and triple negative breast cancer. Analogous to the success of poly (ADP-ribose) polymerase (PARP) inhibitors in BRCA1 and BRCA2 (breast cancer 1 and 2, early onset)-deficient patients that display defects in HR- mediated DNA repair,23 future work to characterize the molec- ular mechanisms of DNA repair in RB1-defective cancers may facilitate the identification of novel therapeutic approaches for patients harboring RB1-negative tumors. Disclosure of potential conflicts of interest No potential conflicts of interest were disclosed. Funding The work was supported through Cancer Research UK (grant C107/10433 and CRUK/A15043 to S.M.) and Worldwide Cancer Research (grant num- ber 12-1280 to S.M.). R.C. was supported by an ICR student fellowship. P. H.H. is supported by the Wellcome Trust (WT089028). References 1. Classon M, Harlow E. The retinoblastoma tumour suppressor in development and cancer. Nat Rev Cancer 2002; 2:910-7; PMID:12459729; http://dx.doi.org/10.1038/nrc950 2. Khidr L, Chen PL. RB, the conductor that orchestrates life, death and differentiation. Oncogene 2006; 25:5210-9; PMID:16936739; http:// dx.doi.org/10.1038/sj.onc.1209612 3. Friend SH, Bernards R, Rogelj S, Weinberg RA, Rapaport JM, Albert DM, Dryja TP. A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 1986; 323:643-6; PMID:2877398; http://dx.doi.org/10.1038/323643a0 4. Morris EJ, Dyson NJ. Retinoblastoma protein partners. Adv Cancer Res 2001; 82:1-54; PMID:11447760 5. Ferreira R, Magnaghi-Jaulin L, Robin P, Harel-Bellan A, Trouche D. The three members of the pocket proteins family share the ability to repress E2F activity through recruitment of a histone deacetylase. Proc Natl Acad Sci U S A 1998; 95:10493-8; PMID:9724731; http://dx. doi.org/10.1073/pnas.95.18.10493 6. Rubin SM, Gall AL, Zheng N, Pavletich NP. Structure of the Rb C-ter- minal domain bound to E2F1-DP1: a mechanism for phosphoryla- tion-induced E2F release. Cell 2005; 123:1093-106; PMID:16360038; http://dx.doi.org/10.1016/j.cell.2005.09.044 7. Lee JO, Russo AA, Pavletich NP. Structure of the retinoblastoma tumour- suppressor pocket domain bound to a peptide from HPV E7. Nature 1998; 391:859-65; PMID:9495340; http://dx.doi.org/10.1038/36038 8. Lee C, Chang JH, Lee HS, Cho Y. Structural basis for the recognition of the E2F transactivation domain by the retinoblastoma tumor sup- pressor. Genes Dev 2002; 16:3199-212; PMID:12502741; http://dx.doi. org/10.1101/gad.1046102 9. Xiao B, Spencer J, Clements A, Ali-Khan N, Mittnacht S, Broceno C, Burghammer M, Perrakis A, Marmorstein R, Gamblin SJ. Crystal structure of the retinoblastoma tumor suppressor protein bound to E2F and the molecular basis of its regulation. Proc Natl Acad Sci U S A 2003; 100:2363-8; PMID:12598654; http://dx.doi.org/10.1073/ pnas.0436813100 10. Talluri S, Dick FA. Regulation of transcription and chromatin struc- ture by pRB: here, there and everywhere. Cell Cycle 2012; 11:3189-98; PMID:22895179; http://dx.doi.org/10.4161/cc.21263 11. Goodrich DW. How the other half lives, the amino-terminal domain of the retinoblastoma tumor suppressor protein. J Cell Physiol 2003; 197:169-80; PMID:14502556; http://dx.doi.org/ 10.1002/jcp.10358 12. Hassler M, Singh S, Yue WW, Luczynski M, Lakbir R, Sanchez-San- chez F, Bader T, Pearl LH, Mittnacht S. Crystal structure of the retino- blastoma protein N domain provides insight into tumor suppression, ligand interaction, and holoprotein architecture. Mol Cell 2007; 28:371-85; PMID:17996702; http://dx.doi.org/10.1016/j. molcel.2007.08.023 13. Cook R, Zoumpoulidou G, Luczynski MT, Rieger S, Moquet J, Spans- wick VJ, Hartley JA, Rothkamm K, Huang PH, Mittnacht S. Direct Involvement of Retinoblastoma Family Proteins in DNA Repair by Non-homologous End-Joining. Cell Rep 2015; 10:2006-18; PMID:25818292; http://dx.doi.org/10.1016/j.celrep.2015.02.059 14. Lieber MR, Ma Y, Pannicke U, Schwarz K. Mechanism and regulation of human non-homologous DNA end-joining. Nat Rev Mol Cell Biol 2003; 4:712-20; PMID:14506474; http://dx.doi.org/10.1038/nrm1202 15. Lieber MR. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem 2010; 79:181-211; PMID:20192759; http://dx.doi.org/10.1146/annurev. biochem.052308.093131 16. Manning AL, Dyson NJ. RB: mitotic implications of a tumour sup- pressor. Nat Rev Cancer 2012; 12:220-6; PMID:22318235 17. Lai A, Lee JM, Yang WM, DeCaprio JA, Kaelin WG, Jr., Seto E, Bran- ton PE. RBP1 recruits both histone deacetylase-dependent and -inde- pendent repression activities to retinoblastoma family proteins. Mol Cell Biol 1999; 19:6632-41; PMID:10490602 18. Gonzalo S, Garcia-Cao M, Fraga MF, Schotta G, Peters AH, Cotter SE, Eguıa R, Dean DC, Esteller M, Jenuwein T, et al. Role of the RB1 fam- ily in stabilizing histone methylation at constitutive heterochromatin. Nat Cell Biol 2005; 7:420-8; PMID:15750587; http://dx.doi.org/ 10.1038/ncb1235 19. Isaac CE, Francis SM, Martens AL, Julian LM, Seifried LA, Erdmann N, Binne UK, Harrington L, Sicinski P, Berube NG, et al. The retino- blastoma protein regulates pericentric heterochromatin. Mol Cell Biol 2006; 26:3659-71; PMID:16612004; http://dx.doi.org/10.1128/ MCB.26.9.3659-3671.2006 20. Carr SM, Munro S, Zalmas LP, Fedorov O, Johansson C, Krojer T, Sagum CA, Bedford MT, Oppermann U, La Thangue NB. Lysine methylation-dependent binding of 53BP1 to the pRb tumor suppres- sor. Proc Natl Acad Sci U S A 2014; 111:11341-6; PMID:25049398; http://dx.doi.org/10.1073/pnas.1403737111 21. Aparicio T, Baer R, Gautier J. DNA double-strand break repair path- way choice and cancer. DNA Repair (Amst) 2014; 19:169-75; PMID:24746645; http://dx.doi.org/10.1016/j.dnarep.2014.03.014 22. Heyer WD, Ehmsen KT, Liu J. Regulation of homologous recombina- tion in eukaryotes. Annu Rev Genet 2010; 44:113-39; PMID:20690856; http://dx.doi.org/10.1146/annurev-genet-051710- 150955 23. Lord CJ, Ashworth A. Targeted therapy for cancer using PARP inhibi- tors. Curr Opin Pharmacol 2008; 8:363-9; PMID:18644251; http://dx. doi.org/10.1016/j.coph.2008.06.016 24. Lamber EP, Beuron F, Morris EP, Svergun DI, Mittnacht S. Structural insights into the mechanism of phosphoregulation of the retinoblas- toma protein. PLoS One 2013; 8:e58463; PMID:23516486; http://dx. doi.org/10.1371/journal.pone.0058463 MOLECULAR CELLULAR ONCOLOGY e1053596-3