ArticlePDF Available

CCDC98 is a BRCA1-BRCT domain-binding protein involved in the DNA damage response

Authors:

Abstract

The product of the breast cancer-1 gene, BRCA1, plays a crucial part in the DNA damage response through its interactions with many proteins, including BACH1, CtIP and RAP80. Here we identify a coiled-coil domain-containing protein, CCDC98, as a BRCA1-interacting protein. CCDC98 colocalizes with BRCA1 and is required for the formation of BRCA1 foci in response to ionizing radiation. Moreover, like BRCA1, CCDC98 has a role in radiation sensitivity and damage-induced G2/M checkpoint control. Together, these results suggest that CCDC98 is a mediator of BRCA1 function involved in the mammalian DNA damage response.
CCDC98 is a BRCA1-BRCT domain–binding protein
involved in the DNA damage response
Hongtae Kim
1,2
, Jun Huang
1,2
& Junjie Chen
1
The product of the breast cancer-1 gene, BRCA1, plays a crucial part in the DNA damage response through its interactions with
many proteins, including BACH1, CtIP and RAP80. Here we identify a coiled-coil domain–containing protein, CCDC98, as a
BRCA1-interacting protein. CCDC98 colocalizes with BRCA1 and is required for the formation of BRCA1 foci in response to
ionizing radiation. Moreover, like BRCA1, CCDC98 has a role in radiation sensitivity and damage-induced G2/M checkpoint control.
Together, these results suggest that CCDC98 is a mediator of BRCA1 function involved in the mammalian DNA damage response.
To survive and maintain their genomic integrity, cells are equipped
with the ability to sense and respond to DNA damage1,2.The
importance of this surveillance system has been demonstrated by
the finding that inactivation of the DNA damage response can lead to
cancer-susceptibility syndromes and neoplastic transformation. Many
proteins, including the protein kinase ataxia-telangiectasia mutated
(ATM), phosphorylated histone H2AX (gH2AX) and mediator of
DNA damage checkpoint-1 (MDC1), are involved in sensing, trans-
ducing and responding to DNA damage signals3. The product of
BRCA1 is also a checkpoint mediator, and its BRCT domains function
in this process by interacting with phosphoserine or phosphothreo-
nine motifs4–6. Previous studies have shown that the BRCA1-BRCT
domains are important for BRCA1’s functions in tumor suppression7
and the DNA damage response8–10. In the presence of DNA lesions,
BRCA1 participates in many DNA damage response pathways, includ-
ing cell-cycle checkpoints during S phase and at the G2/M transition,
and DNA repair via homologous recombination8–11. Defects in these
checkpoints and DNA repair may underlie the increased tumori-
genesis observed in patients with BRCA1 mutations.
Although BRCA1 is known to be recruited to DNA breaks and to
participate in checkpoint regulation, it is not yet clear how the
recruitment of BRCA1 is controlled in the cell. To gain further insights
into the regulation of BRCA1 upon DNA damage, we sought to
identify previously unknown BRCA1-BRCT domain–binding proteins
using a tandem affinity-purification approach. Here we report that
human CCDC98 protein associates with BRCA1 and demonstrate
that CCDC98 acts upstream of BRCA1 and regulates the BRCA1-
dependent DNA damage signaling pathway.
RESULTS
CCDC98 is a BRCA1-associated protein
To identify additional BRCA1-associated proteins, we purified BRCA1-
BRCT domain–containing complexes from human embryonic
kidney 293T cells stably expressing a BRCA1-BRCT domain with
an N-terminal triple tag comprising an S tag, a Flag epitope and a
streptavidin-binding peptide (SFB–BRCA1-BRCT). Mass spectro-
metry revealed a number of known BRCA1-associated proteins,
including BRCA1-associated C-terminal helicase (BACH1), CtBP-
interacting protein (CtIP) and receptor associated protein-80
(RAP80)6,12–15. In the same experiment, we also identified several
putative BRCA1-associated proteins (Supplementary Table 1 online).
Among these, we paid special attention to a coiled-coil domain–
containing protein, CCDC98. This protein contains an SPTF motif at
its extreme C terminus; an identical sequence in BACH1 is required
for interaction of BACH1 with BRCA1-BRCT domains6. The physio-
logical function of CCDC98 is unknown. Notably, we also identified
CCDC98 as a RAP80-associated protein in a tandem affinity purifica-
tion of RAP80-containing complexes (Supplementary Table 2
online), confirming that CCDC98 and RAP80 interact. As both
CCDC98 and RAP80 exist in BRCA1-containing complexes (Supple-
mentary Tables 1 and 2), we speculated that these three proteins
might form a complex.
BRCA1 specifically binds the SPTF motif of CCDC98
We confirmed the association of CCDC98 with BRCA1 and RAP80
using coimmunoprecipitation experiments (Fig. 1a). In addition,
bacterially expressed glutathione S-transferase (GST)-tagged BRCA1-
BRCT domain and GST-RAP80 pulled down CCDC98 from cell
extracts (Fig. 1b), again confirming that CCDC98 interacts with
both BRCA1 and RAP80. Notably, although CCDC98 interacted with
the BRCA1-BRCT domain in a phosphorylation-dependent manner,
its associationwith RAP80 was phosphorylation independent (Fig. 1b).
Prompted by this phosphorylation-dependent interaction between
BRCA1-BRCT and CCDC98, we examined whether the C-terminal
SPTF motif of CCDC98 is required for its interaction with BRCA1-
BRCT. GST–BRCA1-BRCT specifically bound wild-type CCDC98 and
Received 23 April; accepted 26 June; published online 22 July 2007; doi:10.1038/nsmb1277
1
Department of Therapeutic Radiology, Yale University School of Medicine, P.O. Box 208040, New Haven, Connecticut 06520, USA.
2
These authors contributed
equally to this work. Correspondence should be addressed to J.C. (junjie.chen@yale.edu).
710 VOLUME 14 NUMBER 8 A UGUST 2007 NATURE STRUCTURAL & MOLECULAR BIOLOGY
ARTICLES
©2007 Nature Publishing Group http://www.nature.com/nsmb
did not bind CCDC98 lacking the C-terminal SPTF sequence (CCDC
98DSPTF; Fig. 1c). We also generated several point mutations in the
SPTF motif of CCDC98. Whereas GST–BRCA1-BRCT specifically
pulled down wild-type CCDC98, its affinities for the CCDC98 point
mutants were greatly diminished (Fig. 1d). Using a phosphospecific
antibody against the Ser406 residue in the SPTF motif, we confirmed
that this serine residue is indeed phosphorylated in vivo (Fig. 1e). This
phosphorylation and the BRCA1-CCDC98 interaction did not
change after DNA damage (data not shown). Only wild-type
BRCA1, and not a BRCA1 variant lacking the BRCT regions
(BRCA1DBRCT), associated with CCDC98 in vivo (Fig. 1f). Together,
these data suggest that CCDC98 binds BRCA1 in a phosphorylation-
dependent manner through an interaction between BRCA1-BRCTand
the C-terminal SPTF motif of CCDC98.
Figure 1 Identification of CCDC98 as a BRCA1-
binding protein. (a) The interaction between
endogenous CCDC98 and BRCA1 or RAP80.
Immunoprecipitation (IP) reactions were
done using preimmune serum (prebleed) or
anti-CCDC98. Western blotting analyses (W)
were done with indicated antibodies.
(b) Phosphorylation-dependent interaction
between BRCA1-BRCT and CCDC98. GST,
GST–BRCA1-BRCT or GST-RAP80 was incubated
with cell lysates containing exogenously
expressed Flag-tagged wild-type CCDC98, with
or without phosphatase. Bound CCDC98 was
analyzed by anti-Flag immunoblotting. Lower gel
shows amounts of proteins used in these
experiments. (c,d) Beads with GST–BRCA1-BRCT
were incubated with cell lysates containing
exogenously expressed SFB-tagged wild-type
CCDC98, CCDC98DSPTF or SPTF point mutants
with the C-terminal sequences indicated in their
names (CCDC98APTF, CCDC98SATF,
CCDC98SPAF and CCDC98SPTA). Bound
CCDC98 proteins were analyzed by anti-Flag
immunoblotting. (e) CCDC98 is phosphorylated
at Ser406. IP reactions using anti-CCDC98 were
followed by mock or phosphatase treatment. Western blotting was done with indicated antibodies. (f) 293T cells were transfected with plasmid encoding
Myc-BRCA1 or Myc-BRCA1DBRCT and with plasmid encoding SFB-CCDC98. Cell lysates were subjected to immunoprecipitation and immunoblotting with
indicated antibodies (upper blots). Lower blot shows amounts of SFB-tagged CCDC98 in lysates.
a
W : Anti-BRCA1
W : Anti-Flag
(CCDC98)
No PPase
GST-RAP80
GST–BRCA1
BRCT
GST
PPase
treatment
Prebleed
Anti-CCDC98
10% input
GST
GST
GST
–BRCA1-BRCT
GST-RAP80
IP
W : Anti-RAP80
W : Anti-Flag
(CCDC98)
W : Anti-Flag
(CCDC98)
(Cell lysates)
W : Anti-Flag
(CCDC98)
W : Anti-Flag
(CCDC98)
(Cell lysates)
SFB-CCDC98
IP : Anti-Myc
W : Anti-Flag
(CCDC98)
W : Anti-Flag
(CCDC98)
(
Cell l
y
sates
)
W : Anti-Myc
(BRCA1)
W : Anti-CCDC98
c
d
b
SFB-CCDC98
SFB-CCDC98
SPTF
Myc-BRCA1
BRCT
Pull-down :
GST–BRCA1-BRCT
Pull-down :
GST–BRCA1-BRCT
SFB-CCDC98
SFB-CCDC98APTF
SFB-CCDC98SATF
SFB-CCDC98SPAF
SFB-CCDC98SPTA
IP : Anti-CCDC98
PPase :
W : Anti-pS406
W : Anti-CCDC98
–+
fe
Myc-BRCA1
c
d
CCDC98 DAPI
γH2AX
ab
CCDC98
0 Gy
10 Gy
DAPI
γH2AX CCDC98 DAPIBRCA1
CCDC98 siRNA
BRCA1 DAPI
γH2AX
RAP80 DAPI
γH2AX
CCDC98 DAPI
γH2AX
Control siRNA
BRCA1 DAPI
γH2AX
RAP80 DAPI
γH2AX
CCDC98 DAPI
γH2AX
RAP80 siRNA
BRCA1 DAPI
γH2AX
RAP80 DAPI
γH2AX
CCDC98 DAPI
γH2AX
BRCA1 siRNA
BRCA1 DAPI
γH2AX
RAP80
Control siRNA
BRCA1 siRNA
RAP80 siRNA
CCDC98 siRNA
DAPI
γH2AX
W : Anti-BRCA1
W : Anti-RAP80
W : Anti-CCDC98
W : Anti–β-actin
Figure 2 Localization of CCDC98 in cells exposed to ionizing
radiation. (a,b) DNA damage–induced RAP80 focus formation
and colocalization with gH2AX (a) and BRCA1 (b). Mock-
treated or irradiated 293T cells were fixed and stained with
monoclonal antibody to gH2AX or BRCA1, or polyclonal
antibody to CCDC98. (c) Requirement of CCDC98 for
damage-induced BRCA1 focus formation. U2OS cells were
transfected with indicated siRNAs, exposed to ionizing
radiation (10 Gy) and immunostained with monoclonal
antibody to gH2AX or polyclonal antibody to CCDC98,
BRCA1 or RAP80. (d) Western blotting analysis (W) of
BRCA1, RAP80 and CCDC98 expression levels in cells
transfected with indicated siRNAs.
ARTICLES
NATURE STRUCTURAL & MOLECULAR BIOLOGY VOLUME 14 NUMBER 8 AUGUST 2007 711
©2007 Nature Publishing Group http://www.nature.com/nsmb
CCDC98 and BRCA1 form foci after DNA damage
As BRCA1 localizes to sites of DNA breaks in cells exposed to ionizing
radiation, we checked the localization of CCDC98 before and after
DNA damage. Using an antibody to CCDC98, we found the protein to
be evenly distributed in the nucleoplasm of normal cells (Fig. 2a).
After cells were exposed to ionizing radiation, CCDC98 localized to
DNA damage–induced foci and colocalized with gH2AX (a marker of
DNA damage) and BRCA1 (Fig. 2a,b). This indicates that the
localization of CCDC98, like that of BRCA1, is regulated in response
to DNA damage. Notably, we discovered that BRCA1 did not accu-
mulate at DNA breaks in cells where CCDC98 messenger RNA was
depleted using short interfering RNA
(siRNA); however, the localization of RAP80
to damage sites was normal in these cells
(Fig. 2c). Moreover, formation of both
BRCA1 and CCDC98 foci was abolished
in RAP80-depleted cells, but formation of
CCDC98 and RAP80 foci was normal in
BRCA1-depleted cells (Fig. 2c). As a control,
we showed that the expression level of BRCA1
is the same with or without CCDC98 knock-
down (Fig. 2d). In addition, RAP80 knock-
down also does not change the expression of
CCDC98 or BRCA1 (Fig. 2d). Collectively,
these data suggest that CCDC98 acts down-
stream of RAP80 but upstream of BRCA1 in
the DNA damage response pathway.
CCDC98 focus formation depends on its
N terminus
Our results suggested that CCDC98 forms a
complex with RAP80 and BRCA1 and loca-
lizes to sites of damaged DNA. Next, we
attempted to determine which regions of
CCDC98 are important for its localiza-
tion to foci. Full-length CCDC98 and
CCDC98DSPTF mutant localized normally
to nuclear foci in cells with DNA damage, whereas all of the other
CCDC98 deletion mutants we tested did not (Fig. 3a). All three
N-terminal and internal deletion mutants of CCDC98 also did
not bind RAP80 (Fig. 3b), whereas CCDC98DSPTF and a
CCDC98 mutant with a large C-terminal deletion (CCDC98D4)
were defective in BRCA1 binding (Fig. 3c). Because it localizes
to the cytoplasm, it is difficult to interpret the results obtained
with the CCDC98D4 mutant (two putative nuclear localization
sequences, 358-Lys-Arg-Ser-Arg-361 and 368-Lys-Arg-Ser-Lys-371,
are deleted in this mutant). Nevertheless, these data suggest that
CCDC98 mediates the interaction between BRCA1 and RAP80
CCDC98WT
CCDC98D1
CCDC98D1
CCDC98D2
CCDC98D2
CCDC98D3
CCDC98D3
CCDC98D4
CCDC98D4CCDC98WT
DAPI
Pull-down : GST-RAP80 Pull–down : GST–BRCA1-BRCT
Flag
(CCDC98)
γH2AX
CCDC98SPTF
CCDC98
SPTF
1
100
101 200
201
250
300
405
409
Focus
formation
BRCA1
binding
RAP80
binding
+
+
?
+
+
+
+
+
+
+
SFB-CCDC98D4
SFB-CCDC98
SFB-CCDC98D2
SFB-CCDC98D3
SFB-CCDC98D1
SFB-CCDC98
SPTF
SFB-CCDC98D4
SFB-CCDC98
SFB-CCDC98D2
SFB-CCDC98D3
SFB-CCDC98D1
SFB-CCDC98
SPTF
SFB-RAP80WT
SFB-RAP80D1
SFB-RAP80D2
SFB-RAP80D3
SFB-RAP80D4
SFB-RAP80D5
SFB-RAP80D6
SFB-RAP80WT
SFB-RAP80D1
SFB-RAP80D2
SFB-RAP80D3
SFB-RAP80D4
SFB-RAP80D5
SFB-RAP80D6
a
bc
W : Anti-Flag
(CCDC98)
W : Anti-Flag
(CCDC98)
(Cell lysates)
W : Anti-Flag
(CCDC98)
W : Anti-Flag
(CCDC98)
(Cell lysates)
dRAP80WT UIM UIM ZF ZF
719
CCDC9
8
binding
RAP80D1
RAP80D2
RAP80D3
RAP80D4
RAP80D5
RAP80D6
1
+
+
+
+
+
+
71
93
127
200
235 337
336 491
470 583
582
IP :
Anti-Flag
W : Anti-Myc
(CCDC98)
W : Anti-Flag
(RAP80)
W : Anti-Myc
(CCDC98)
W : Anti-Flag
(RAP80)
Whole-cell
lysates
Whole–cell
lysates
W : Anti-Flag
(RAP80)
W : Anti-Myc
(CCDC98)
W : Anti-Flag
(RAP80)
W : Anti-Myc
(
CCDC98
)
IP :
Anti-Myc
Figure 3 Focus localization of CCDC98 depends
on its N-terminal RAP80-binding region.
(a) 293T cells were transfected with SFB-tagged
wild-type (WT) CCDC98 or deletion mutants
shown in diagram. After 24 h, cells were exposed
to 10 Gy of ionizing radiation. Eight hours after
irradiation, cells were fixed and stained with
monoclonal anti-Flag or polyclonal anti-gH2AX.
(b,c) Mapping of the RAP80- and BRCA1-
interacting domains in CCDC98. Beads coated
with GST-RAP80 (b) or GST–BRCA1-BRCT (c)
were incubated with cell lysates containing
exogenously expressed SFB-tagged WT CCDC98
or deletion mutants. After extensive washing,
bound RAP80 was analyzed by western
blotting (W) with anti-Flag. (d) SFB-tagged WT
RAP80 and its internal deletion mutants were
used to map the CCDC98-interacting domain in
RAP80. 293T cells were transfected with
plasmids encoding Myc-CCDC98 and the
indicated SFB-RAP80 proteins. Cell lysates were
subjected to immunoprecipitation (IP) and
immunoblotting with indicated antibodies
(top blots). Bottom blots show amounts of
SFB-RAP80 and Myc-CCDC98 in these lysates.
ARTICLES
712 VOLUME 14 NUMBER 8 A UGUST 2007 NATURE STRUCTURAL & MOLECULAR BIOLOGY
©2007 Nature Publishing Group http://www.nature.com/nsmb
and that the localization of CCDC98 to foci depends on its interaction
with RAP80.
We confirmed a strong interaction between CCDC98 and RAP80
using a baculovirus-insect cell system (Supplementar y Fig. 1 online).
Using a series of deletion mutants of RAP80, we identified a region
(residues 235–337) on the C-terminal side of the ubiquitin-interacting
motifs (UIMs) that is required for its interaction with CCDC98
(Fig. 3d). The same region of RAP80 is also important for its
association with BRCA1 in vivo (Supplementary Fig. 2 online), an
observation which agrees with our proposal that CCDC98 bridges the
interaction between RAP80 and BRCA1.
CCDC98 is required in the G2/M checkpoint
The loss of BRCA1 leads to defects in the DNA damage response—in
particular, impaired cell-cycle checkpoints and increased sensitivity to
DNA damaging agents16,17. We therefore examined whether the loss
of CCDC98 results in similar defects in the DNA damage response.
Both CCDC98 siRNAs we synthesized efficiently decreased CCDC98
expression in cells (Fig. 4a). Cells treated with these siRNAs showed
defective G2/M checkpoint control after DNA damage (Fig. 4b and
Supplementary Fig. 3 online). The protein kinase CHK1 acts down-
stream of BRCA1 and is required for this G2/M checkpoint control
in response to ionizing radiation18–21. If CCDC98 functions upstream
of BRCA1, a defect in CHK1 activation is expected in cells depleted
of CCDC98. This is indeed the case (Fig. 4c). CCDC98 knockdown
cells were also more sensitive to radiation than cells transfected
with control siRNA (Fig. 4d). These data indicate that CCDC98 is a
key upstream regulator that influences BRCA1 function upon DNA
damage (Fig. 4e).
DISCUSSION
In this study, we identified CCDC98 as a BRCA1-BRCT–binding
protein. Like BRCA1, CCDC98 normally exists in the nucleoplasm but
localizes to DNA breaks after exposure to ionizing radiation. CCDC98
also participates in the BRCA1-dependent G2/M checkpoint control,
suggesting that CCDC98 functions together with BRCA1 in the DNA
damage response.
Besides CCDC98, the ubiquitin-interacting protein RAP80 was also
identified as a BRCA1-associated protein in our biochemical purifica-
tion of BRCA1-containing complexes. Studies from several groups,
including ours, have demonstrated that RAP80 acts upstream of
BRCA1 and regulates BRCA1 localization and function after DNA
damage13–15. Moreover, another group has also identified CCDC98
(called Abraxas in their study) as a BRCA1-interacting protein15.
Similar to our current study, they also showed that CCDC98/Abraxas
interacts with BRCA1 in a phosphorylation-dependent manner via its
C-terminal SPTF motif15. Here, we have expanded on our initial
observations and demonstrated a hierarchy in this DNA damage signal
transduction pathway. We show that although RAP80 is required for
formation of both CCDC98 and BRCA1 foci, CCDC98 is required for
formation of only BRCA1 and not RAP80 foci. Moreover, abolishing
BRCA1 does not affect either RAP80 or CCDC98 focus formation
after DNA damage. Thus, we are able to delineate a signaling pathway
from RAP80 to CCDC98 and then to BRCA1 (Fig. 4e).
Our study also permits a better understanding of CCDC98’s activity
as a mediator in this process. We show that the N terminus of
CCDC98 is required for RAP80 binding, and its C-terminal phos-
phorylation motif is required for BRCA1 binding. In agreement with
the notion that CCDC98 functions downstream of RAP80, only the
N-terminal RAP80-binding domain of CCDC98 is important for its
localization to foci after DNA damage. Putting all these studies
together, we now have a better understanding of the mechanisms
underlying the recruitment of BRCA1 to damaged DNA. RAP80
binds directly to the N terminus of CCDC98. This interaction is not
phosphorylation dependent, but rather allows formation of a
stable complex between RAP80 and CCDC98. After DNA damage,
the RAP80–CCDC98 complex localizes to damage sites. RAP80’s
localization to foci depends on its UIM domain, which probably
binds unidentified ubiquitinated proteins at DNA breaks. Through
its C-terminal phosphorylation motif, CCDC98 then recruits
BRCA1 to the DNA damage sites and regulates BRCA1-dependent
checkpoint control.
ab
c
e
d
W : Anti–β-actin
W : Anti–β-actin
W : Anti-CCDC98
Control
siRNA
Control
siRNA
CCDC98
siRNA2
CCDC98
siRNA2
BRCA1
siRNA
RAP80
siRNA
CCDC98
siRNA1
CCDC98
siRNA1
CCDC98
siRNA1
3.0
2.5
2.0
1.5
Percentage of mitotic cells
1.0
0.5
0
0 Gy
2 Gy
CCDC98 siRNA
Control siRNA
100
80
60
Surviving colonies (%)
40
20
0
0
12345
Radiation (Gy)
Control
siRNA
10 Gy :
W : Anti-pCHK1
W : Anti-CHK1
W : Anti-CCDC98
––++
IR
ATM
pH2AX/MDC1
E3 ligase ?
XUb
RAP80
CCDC98
BRCA1
Y ?
DNA
re
p
air?
Damage
checkpoint
Ub
Figure 4 Requirement of CCDC98 for ionizing radiation–induced DNA
damage response. (a) Western blotting analysis (W) of CCDC98 expression
in cells transfected with indicated siRNAs. (b) G2/M checkpoint control in
CCDC98 knockdown cells. HeLa cells transfected with indicated siRNAs
were exposed to 0 or 2 Gy of ionizing radiation. Cells were fixed and stained
with histone-specific anti-pH3 (a mitotic marker) and propidium iodide.
Percentages of mitotic cells were determined by FACS analysis. Data shown
are averages of three independent experiments; error bars indicate s.d.
(c) Requirement of CCDC98 for CHK1 phosphorylation after DNA damage.
Control or CCDC98 siRNA–transfected HeLa cells were exposed to 0 or
10 Gy of ionizing radiation, harvested 2 h later and immunoblotted with
indicated antibodies. (d) Radiation sensitivity of cells lacking CCDC98. HeLa
cells were transfected with control or CCDC98 siRNAs. Cells were irradiated
with indicated doses of ionizing radiation. Percentage of colonies surviving
was determined 10–12 d later. Experiments were done in triplicate; results
shown are averages of two or three independent experiments at each dose;
error bars indicate s.d. (e) Model of the DNA damage response pathway that
integrates CCDC98. Our data indicate that CCDC98 operates upstream of
BRCA1 and specifically regulates BRCA1 localization and function after
DNA damage.
ARTICLES
NATURE STRUCTURAL & MOLECULAR BIOLOGY VOLUME 14 NUMBER 8 AUGUST 2007 713
©2007 Nature Publishing Group http://www.nature.com/nsmb
The factors that act upstream of the RAP80–CCDC98 complex and
recruit it to sites of DNA damage remain elusive. What we do know is
that both gH2AX and MDC1 are upstream regulators and are required
for the focus formation of many checkpoint proteins, including
RAP80 and BRCA1. Because the localization of RAP80 seems to
depend on the ability of its UIM domain to bind polyubiquitinated
proteins, we speculate that there is at least one E3 ubiquitin ligase
involved in this process. This unidentified E3 ligase may act after
gH2AX and MDC1 to facilitate protein ubiquitination at sites of DNA
damage, which would in turn serve as a signal to recruit RAP80–
CCDC98 and BRCA1. The identification of this E3 ligase and its
substrates at DNA breaks would provide further insight into the
complex regulation of DNA damage response pathways.
Although RAP80 and CCDC98 seem to function upstream of
BRCA1 in the DNA damage signal transduction pathway, it is
noteworthy that checkpoint defects observed in RAP80-or
CCDC98-deficient cells are not as severe as those observed in cells
with a BRCA1 deficiency. One likely explanation is that there are
proteins other than RAP80 and CCDC98 that also participate in
regulating BRCA1 function after DNA damage. We hope that future
studies will identify this parallel pathway, revealing exactly how the
tumor suppressor BRCA1 is regulated after DNA damage and con-
tributes to the maintenance of genomic stability.
METHODS
Plasmids. Human CCDC98 full-length complementary DNA was obtained
using reverse-transcription PCR. Wild-type human CCDC98 and its point
mutants and deletion mutants were generated by PCR and subcloned into a
modified pIRES-EGFP mammalian expression vector to create constructs
encoding SFB-tagged wild-type or mutant CCDC98. DNA fragments encoding
BRCA1-BRCT domain and RAP80 were also generated by PCR and subcloned
into pGEX-4T-1 vector (Pharmacia) to make constructs for expression of GST–
BRCA1-BRCTand GST-RAP80, respectively. Myc-BRCA1, Myc-BRCA1DBRCT,
and full-length human RAP80 and its deletion mutants were described13.
Cell culture and treatment with ionizing radiation. HeLa, U2OS and 293T
cells were purchased from the American Type Culture Collection and main-
tained in RPMI 1,640 medium supplemented with 10% (v/v) FBS at 37 1C
in 5% CO
2
. Cells were irradiated at the indicated doses using a JL Shepherd
137
Cs radiation source. The irradiated cells were then returned to the same
culture conditions and maintained for the periods of time specified in the
figure legends.
Short interfering RNA. All siRNA duplexes used in this study were purchased
from Dharmacon. The sequences of RAP80 siRNA, CCDC98 siRNA 1, CCDC98
siRNA 2, BRCA1 siRNA and the control siRNA are 5¢-GAAGGAUGUGGAAA
CUACCdTdT-3¢,5¢-CAGGGUACCUUUAGUGGUUUU-3¢,5¢-ACACAAGA
CAAACGAUCUAUU-3¢and 5¢-UCACAGUGUCCUUUAUGUAdTdT-3¢and
5¢-UUCAAUAAAUUCUUGAGGUUU-3¢, respectively. siRNAs were transfected
into the cells using Oligofectamine (Invitrogen) according to the manufac-
turer’s instructions.
Antibodies, transfection and immunoprecipitation procedures. Rabbit anti-
bodies to BRCA1, CCDC98 and RAP80 were raised by immunizing rabbits
with GST-BRCA1 fragments, GST-CCDC98 and GST-RAP80 (residues 1–354)
respectively. Phosphospecific antibody to Ser406 was generated by immunizing
rabbits with KLH-conjugated GFGEYSR-pS-PTF peptide. The resulting rabbit
polyclonal sera were affinity-purified using the SulfoLink or AminoLink Plus
Immobilization and Purification Kit (Pierce). gH2AX antibodies were
described22.AntibodiestoFlagandb-actin were purchased from Sigma.
Antibody to phosphorylated histone H3 (pH3) was purchased from Upstate
Biotechnology. Transient transfection was done using Fugene 6 transfection
reagent (Roche) according to the manufacturer’s instructions. For immuno-
precipitation, cells were washed with ice-cold PBS and then lysed in NETN
buffer (0.5% (v/v) Nonidet P-40, 20 mM Tris (pH 8.0), 50 mM NaCl, 50 mM
NaF, 100 mMNa
3
VO
4
, 1 mM DTT and 50 mgml
–1
PMSF) at 4 1C for 10 min.
Crude lysates were cleared by centrifugation at 14,000 r.p.m. (Micro 240A,
Scientific) and 4 1C for 5 min, and supernatants were incubated with protein
A–agarose–conjugated primary antibodies. The immunocomplexes were
washed three times with NETN buffer and then subjected to SDS-PAGE.
Western blotting was done using the antibodies specified in the figures.
Cell lines and affinity purification of SFB-tagged protein complexes. To
establish cell lines stably expressing various epitope-tagged proteins, 293T cells
were transfected with plasmids encoding SFB-tagged proteins and pGK-Puro.
Forty-eight hours after transfection, the cells were split at a 1:10 ratio and
cultured in medium containing puromycin (10 mgml
–1
) for 3 weeks. The
individual puromycin-resistant colonies were isolated and screened by western
blotting. 293T cells stably expressing tagged proteins were lysed with 4 ml
NETN buffer on ice for 10 min. Crude lysates were cleared by centrifugation at
14,000 r.p.m. (Micro 240A, Scientific) at 4 1C for 10 min, and supernatants
were incubated with 300 ml streptavidin-conjugated beads (Amersham). The
immunocomplexes were washed three times with NETN buffer and then bead-
bound proteins were eluted with 500 ml NETN buffer containing 2 mg ml
–1
biotin (Sigma). The eluted supernatant was incubated with 60 ml S protein
beads (Novagen). The immunocomplexes were washed three times with NETN
buffer and subjected to SDS-PAGE. Protein bands were visualized by silver
staining, excised and digested, and the peptides were analyzed by mass
spectrometry.
Glutathione S-transferase pull-down assay. GST fusion protein was expressed
in Escherichia coli and purified as described23. GST fusion protein or GSTalone
(2 mg) was immobilized on glutathione-Sepharose 4B beads and incubated for
2 h at 4 1C with lysates prepared from cells transiently transfected with
plasmids encoding the indicated proteins. After washing with NETN buffer,
the samples were separated by SDS-PAGE and analyzed by western blotting.
Immunofluorescent staining. Cells grown on coverslips were fixed with
3% (w/v) paraformaldehyde at room temperature for 15 min and then
permeabilized with PBS containing 0.5% (v/v) Triton X-100 at room tempera-
ture for 5 min. The coverslips were blocked with PBS containing 5% (v/v) goat
serum at room temperature for 30 min before incubation with primary
antibodies at room temperature for 20 min. After washing with PBS, cells
were incubated with the secondary antibodies fluorescein isothiocyanate–
conjugated goat anti-mouse IgG, rhodamine-conjugated goat anti-rabbit IgG
or rhodamine-conjugated goat anti-mouse IgG (Jackson ImmunoResearch) at
room temperature for 20 min. Nuclei were counterstained with 4,6-diamidino-
2-phenylindole (DAPI). After a final wash with PBS, coverslips were mounted
with glycerin containing p-phenylenediamine. All images were obtained with a
Nikon ECLIPSE E800 fluorescence microscope.
G2/M cell-cycle checkpoint assay. HeLa cells in a 100-mm plate were
transfected twice with control or CCDC98 siRNAs at 24-h intervals. Forty-
eight hours after the second transfection, transfected cells were mock-treated or
irradiated at indicated doses using a JL Shepherd
137
Cs radiation source. One
hour after irradiation, cells were fixed with 70% (v/v) ethanol at –20 1Cfor
24 h, then stained with rabbit antibody to pH3 and incubated with fluorescein
isothiocyanate–conjugated goat secondary antibody to rabbit immunoglobulin.
The stained cells were treated with RNase A, incubated with propidium iodide
and then analyzed by flow cytometry.
Cell survival assays. HeLa cells in a 60-mm plate were transfected twice with
control or CCDC98 siRNAs at 24-h intervals. Forty-eight hours after the second
transfection, transfected cells were irradiated at the indicated doses using a JL
Shepherd
137
Cs radiation source. Ten to twelve days after irradiation, cells were
washed with PBS, fixed and stained with 2% (w/v) methylene blue, and the
colonies were counted.
Note: Supplementary information is available on the Nature Structural &Molecular
Biology website.
ACKNOWLEDGMENTS
We thank members of the Chen laboratory for helpful discussions and technical
support. This work was supported in part by grants from the US National
ARTICLES
714 VOLUME 14 NUMBER 8 A UGUST 2007 NATURE STRUCTURAL & MOLECULAR BIOLOGY
©2007 Nature Publishing Group http://www.nature.com/nsmb
Institute of Health (RO1CA089239 to J.C.) and the US Department of Defense
breast cancer Era of Hope Scholar Award (W81XWH-05-1-0470 to J.C.).
AUTHOR CONTRIBUTIONS
H.K., J.H. and J.C. designed experiments and interpreted the data; H.K. and J.H.
performed all experiments; H.K. and J.C. prepared the manuscript.
COMPETING INTERESTS STATEMENT
The authors declare no competing financial interests.
Published online at http://www.nature.com/nsmb/
Reprints and permissions information is available online at http://npg.nature.com/
reprintsandpermissions
1. Zhou, B.B. & Elledge, S.J. The DNA damage response: putting checkpoints in
perspective. Nature 408, 433–439 (2000).
2. Rouse, J. & Jackson, S.P. Interfaces between the detection, signaling, and repair of
DNA damage. Science 297, 547–551 (2002).
3. Su, T.T. Cellular responses to DNA damage: one signal, multiple choices. Annu. Rev.
Genet. 40, 187–208 (2006).
4. Manke, I.A., Lowery, D.M., Nguyen, A. & Yaffe, M.B.BRCT repeats as phosphopeptide-
binding modules involved in protein targeting. Science 302, 636–639 (2003).
5. Yu, X., Chini, C.C., He, M., Mer, G. & Chen, J. The BRCT domain is a phospho-protein
binding domain. Science 302, 639–642 (2003).
6. Yu, X. & Chen, J. DNA damage-induced cell cycle checkpoint control requires CtIP, a
phosphorylation-dependent binding partner of BRCA1 C-terminal domains. Mol. Cell.
Biol. 24, 9478–9486 (2004).
7. Ludwig, T., Fisher, P., Ganesan, S. & Efstratiadis, A. Tumorigenesis in mice carrying a
truncating Brca1 mutation. Genes Dev. 15, 1188–1193 (2001).
8. Scully, R. & Livingston, D.M. In search of the tumour-suppressor functions of BRCA1
and BRCA2. Nature 408, 429–432 (2000).
9. Deng, C.X. & Brodie, S.G. Roles of BRCA1 and its interacting proteins. Bioessays 22,
728–737 (2000).
10. Venkitaraman, A.R. Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell
108, 171–182 (2002).
11. Deng, C.X. Roles of BRCA1 in centrosome duplication. Oncogene 21, 6222–6227
(2002).
12. Cantor, S.B. et al. BACH1, a novel helicase-like protein, interacts directly with BRCA1
and contributes to its DNA repair function. Cell 105, 149–160 (2001).
13. Kim, H., Chen, J. & Yu, X. Ubiquitin-binding protein RAP80 mediates BRCA1-
dependent DNA damage response. Science 316, 1202–1205 (2007).
14. Sobhian, B. et al. RAP80 targets BRCA1 to specific ubiquitin structures at DNA
damage sites. Science 316, 1198–1202 (2007).
15. Wang, B. et al. Abraxas and RAP80 form a BRCA1 protein complex required for the
DNA damage response. Science 316, 1194–1198 (2007).
16. Xu, B., Kim, S. & Kastan, M.B. Involvement of Brca1 in S-phase and G
2
-phase
checkpoints after ionizing irradiation. Mol. Cell. Biol. 21, 3445–3450 (2001).
17. Cortez, D., Wang, Y., Qin, J. & Elledge, S.J. Requirement of ATM-dependent phosphor-
ylation of brca1 in the DNA damage response to double-strand breaks. Science 286,
1162–1166 (1999).
18. Liu, Q. et al. Chk1 is an essential kinase that is regulated by Atr and required for the
G
2
/M DNA damage checkpoint. Genes Dev. 14, 1448–1459 (20 00).
19. Takai, H. et al. Aberrant cell cycle checkpoint function and early embryonic death in
Chk1(/)mice.Genes Dev. 14, 1439–1447 (2000).
20. Yarden, R.I., Pardo-Reoyo, S., Sgagias, M., Cowan, K.H. & Brody, L.C. BRCA1
regulates the G2/M checkpoint by activating Chk1 kinase upon DNA damage. Nat.
Genet. 30, 285–289 (2002).
21. Deming, P.B. et al. The human decatenation checkpoint. Proc. Natl. Acad. Sci. USA
98, 12044–12049 (2001).
22. Lou, Z. et al. MDC1 maintains genomic stability by participating in the amplification of
ATM-dependent DNA damage s ignals. Mol. Cell 21, 187–200 (2006 ).
23. Hofer, B., Backhaus, S. & Timmis, K.N. The biphenyl/polychlorinated biphenyl-
degradation locus (bph) of Pseudomonas sp. LB400 encodes four additional metabolic
enzymes. Gene 144,916(1994).
ARTICLES
NATURE STRUCTURAL & MOLECULAR BIOLOGY VOLUME 14 NUMBER 8 AUGUST 2007 715
©2007 Nature Publishing Group http://www.nature.com/nsmb
... While PALB2 serves HR by bridging the coiled-coil domain of BRCA1 with BRCA2 [4,5], BARD1 interacts with BRCA1´s RING domain to aid BRCA1 in counteracting 53BP1 on DNA ends [6]. Through its BRCA1 C-Terminal (BRCT) domains, BRCA1 forms mutually exclusive complexes with the pSPxF-motifs containing proteins ABRAXAS1 (alias ABRA1, CCDC98, FAM175A) [7][8][9], BRIP1 (BRCA1 interacting protein C-terminal helicase 1, alias BACH1, FANCJ) [10,11] and CTIP (C-terminal binding protein-interacting protein, alias RBBP8) [12,13]. These three CDK-mediated, phosphorylation-dependent interactions result in formation of BRCA1-A to -C complexes in Sand G2-phase of the cell cycle [14]. ...
... In BRCA1-C, CTIP and MRE11-RAD50-NBS1 (MRN) initiate end-resection at DSBs and therefore HR [21,22]. There has been a debate on whether BRCA1-A is necessary to ensure proper execution of HR [7,9,15,18,23] or to prevent excessive HR through antagonizing DSB end-resection [24,25]. Nevertheless, there is a general consensus on that the localization of the BRCA1-A complex at DNA damage sites is crucial for repair, DDR and genome stability. ...
... ABRAXAS1 is known to bind to the BRCT repeats in BRCA1 via its extreme C-terminal SPTF motif when phosphorylated post-IR by ATM [7][8][9]50]. BRCA1 exerts key functions in HR that are lost in pathogenic BRCA1 variants mutated in the BRCT domain [51]. In our preceding studies engaging cells from heterozygous carriers of pathogenic BRCA1-, BRCA2-or PALB2-mutations, HR was found to be unperturbed [31,34], while complete loss-of-function was noticeable in bi-allelic carriers of BRCA1-or BRCA2-mutations [34,35]. ...
Article
Full-text available
It has been well-established that mutations in BRCA1 and BRCA2 , compromising functions in DNA double-strand break repair (DSBR), confer hereditary breast and ovarian cancer risk. Importantly, mutations in these genes explain only a minor fraction of the hereditary risk and of the subset of DSBR deficient tumors. Our screening efforts identified two truncating germline mutations in the gene encoding the BRCA1 complex partner ABRAXAS1 in German early-onset breast cancer patients. To unravel the molecular mechanisms triggering carcinogenesis in these carriers of heterozygous mutations, we examined DSBR functions in patient-derived lymphoblastoid cells (LCLs) and in genetically manipulated mammary epithelial cells. By use of these strategies we were able to demonstrate that these truncating ABRAXAS1 mutations exerted dominant effects on BRCA1 functions. Interestingly, we did not observe haploinsufficiency regarding homologous recombination (HR) proficiency (reporter assay, RAD51-foci, PARP-inhibitor sensitivity) in mutation carriers. However, the balance was shifted to use of mutagenic DSBR-pathways. The dominant effect of truncated ABRAXAS1 devoid of the C-terminal BRCA1 binding site can be explained by retention of the N-terminal interaction sites for other BRCA1-A complex partners like RAP80. In this case BRCA1 was channeled from the BRCA1-A to the BRCA1-C complex, which induced single-strand annealing (SSA). Further truncation, additionally deleting the coiled-coil region of ABRAXAS1, unleashed excessive DNA damage responses (DDRs) de-repressing multiple DSBR-pathways including SSA and non-homologous end-joining (NHEJ). Our data reveal de-repression of low-fidelity repair activities as a common feature of cells from patients with heterozygous mutations in genes encoding BRCA1 and its complex partners.
... DNA primers used for PCR, cloning, and sequencing were obtained from macrogen (Seoul, South Korea) and listed in the Supplementary Table S1. The GFP-TRAIP and Myc-TRAIP, TRAIP D-1, D-2, D-3, D-4 and D-5 deletion mutant expression plasmids were previously described (34,35). ZNF212 gene was purchased from the Korea Human Gene Bank. ...
... A list of all siRNA duplexes used in this work can be found in the Supplementary Table S1. Control and TRAIP siRNA were previously described (34,35). The sequences of ZNF212 siRNA used in this work are listed in the Supplementary Table S1. ...
... Anti-TRAIP, -GFP and -␥ H2AX antibodies were previously described (34,35). Other antibodies were purchased as the followings: anti-Flag-HRP (Sigma, A8592), anti-Myc-HRP antibody (Roche, 11814150001), anti-GFP (Clontech, 632380), anti-␤-actin antibody (Sigma, A5441), anti-␣-tubulin antibody (Millopore, 05-829), anti-ZNF212 antibody (Altas antibodies, HPA049807), anti-TRAIP antibody (30), anti-RAD51 antibody (Abcam, ab3801), anti-RPA2 antibody (Bethyl laboratories, A300-244A), anti-53BP1 (Cell signaling technology, #4937), anti-BRCA1 antibody (Santa cruz, SC-6954), anti-FANCD2 antibody (Novus Biologicals, NB100-182), anti-Histone 3 antibody (Santa cruz, sc-8654), anti-NEIL3 antibody (Proteintch, 11621-1-AP) and anti-MCM7 antibody (Santa cruz, sc-9966). ...
Article
Full-text available
TRAIP is a key factor involved in the DNA damage response (DDR), homologous recombination (HR) and DNA interstrand crosslink (ICL) repair. However, the exact functions of TRAIP in these processes in mammalian cells are not fully understood. Here we identify the zinc finger protein 212, ZNF212, as a novel binding partner for TRAIP and find that ZNF212 colocalizes with sites of DNA damage. The recruitment of TRAIP or ZNF212 to sites of DNA damage is mutually interdependent. We show that depletion of ZNF212 causes defects in the DDR and HR-mediated repair in a manner epistatic to TRAIP. In addition, an epistatic analysis of Zfp212, the mouse homolog of human ZNF212, in mouse embryonic stem cells (mESCs), shows that it appears to act upstream of both the Neil3 and Fanconi anemia (FA) pathways of ICLs repair. We find that human ZNF212 interacted directly with NEIL3 and promotes its recruitment to ICL lesions. Collectively, our findings identify ZNF212 as a new factor involved in the DDR, HR-mediated repair and ICL repair though direct interaction with TRAIP.
... In recent years, the expression of CCDC proteins have been demonstrated to be associated malignant tumor tissues and tumor cell lines [34][35][36][37]. CCDC115 has been shown to be related to Golgi trafficking and protein glycosylation, and its deficiency would lead to congenital glycosylation disorder [38,39]. ...
Article
Full-text available
Liver hepatocellular carcinoma (LIHC) is a major malignant tumor of the digestive system with a high incidence rate and poor early diagnosis. Coiled-coil domain-containing protein 115 (CCDC115), an accessory component of vacuolar-ATPase with dramatically abnormal expression, is associated with survival outcomes of cancer patients. However, the role of CCDC115 in LIHC remains unclear. In this study, we aimed to determine the functional role of CCDC115 in LIHC by examining CCDC115 expression, and its influence on LIHC prognosis. Through extensive statistical analyses, using LIHC patient databases, we observed that CCDC115 expression significantly increased in tumor tissues of LIHC patients. In addition, CCDC115 expression correlated with the poor prognosis. Additionally, CCDC115 was found to be involved in several cancer-related pathways, specifically the PI3K-Akt pathway. The expression of CCDC115 was positively correlated with human leukocyte antigen molecules as well as with immune checkpoint molecules in LIHC patients. We performed in vitro experiments and confirmed that the expression of CCDC115 significantly affects the proliferation potential, metastasis and sorafenib resistance of liver cancer cells, as well as some key protein expression in PI3K-Akt pathway. These results indicate that CCDC115 could serve as a diagnostic and prognostic biomarker of LIHC, and targeting CCDC115 may provide a potential strategy to enhance the efficacy of liver cancer therapy.
... Coiled-coil domain is a specific structural protein involved in several pathophysiological processes [5,6]. Coiled-coil domain-containing (CCDC) proteins are expressed abnormally and serve as cancer promoters in multiple malignancies [7][8][9][10][11][12][13]. CCDC3, also known as fat/vessel-derived secretory protein (Favine) and encoded by CCDC3 (NCBI: NP_ 028804), is highly conserved among species [14]. ...
Article
Full-text available
Introduction: Epithelial ovarian cancer (EOC) is a highly aggressive disease whose unique metastatic site is the omentum. Coiled-coil domain containing 3 (CCDC3) is an adipocyte-derived secreted protein that is specifically elevated in omental adipose tissue. However, its function is still unknown. Material and methods: Initially, a Kaplan-Meier plot was applied to evaluate the prognostic value of CCDC3 expression in patients with EOC. A bioinformatics analysis was next used to explore the biological function of CCDC3 in EOC. Western blot, quantitative real-time polymerase chain reaction, and in vitro invasion and migration assays were performed using SKOV3 cells and CCDC3 secreted by rat adipocytes to analyzes the impact of CCDC3 on EOC and the underlying mechanism. Results: Overexpression of CCDC3 was associated with poor prognosis of EOC. CCDC3 interacted with multiple key signalling pathways, including the Wnt/β-catenin pathway. EOC cellular proliferation, migration, and invasion were promoted in vitro when co-cultured with CCDC3 enriched conditioned medium, and this tumour-promoting effect was induced by activating the Wnt/β-catenin pathway. Furthermore, the epithelial-mesenchymal transition of EOC cells was reversed after CCDC3 silencing. Conclusions: Our results support that CCDC3 promotes EOC tumorigenesis through the Wnt/β-catenin pathway and that CCDC3 may serve as a novel prognostic biomarker and therapeutic target for metastatic EOC.
... Transient transfection of plasmid DNA and siRNAs was performed using Lipofectamine 3000 (Thermo Fisher Scientific) and Lipofectamine RNAiMAX (Thermo Fisher Scientific). The control siRNAs were previously described ( 33 ). The following custom siRNA sequences, RPA32 #1: 5 -GGCUCCAA CCAA CAUUGUU-3 and RPA32 #2: 5 -CCUAGUUUCACAAUCUGUU-3 , were synthesized by Bioneer Inc. (South Korea). ...
Article
Full-text available
Replication protein A (RPA), a eukaryotic single-stranded DNA (ssDNA) binding protein, dynamically interacts with ssDNA in different binding modes and plays essential roles in DNA metabolism such as replication, repair, and recombination. RPA accumulation on ssDNA due to replication stress triggers the DNA damage response (DDR) by activating the ataxia telangiectasia and RAD3-related (ATR) kinase, which phosphorylates itself and downstream DDR factors, including RPA. We recently reported that the N-methyl-D-aspartate receptor synaptonuclear signaling and neuronal migration factor (NSMF), a neuronal protein associated with Kallmann syndrome, promotes RPA32 phosphorylation via ATR upon replication stress. However, how NSMF enhances ATR-mediated RPA32 phosphorylation remains elusive. Here, we demonstrate that NSMF colocalizes and physically interacts with RPA at DNA damage sites in vivo and in vitro. Using purified RPA and NSMF in biochemical and single-molecule assays, we find that NSMF selectively displaces RPA in the more weakly bound 8- and 20-nucleotide binding modes from ssDNA, allowing the retention of more stable RPA molecules in the 30-nt binding mode. The 30-nt binding mode of RPA enhances RPA32 phosphorylation by ATR, and phosphorylated RPA becomes stabilized on ssDNA. Our findings provide new mechanistic insight into how NSMF facilitates the role of RPA in the ATR pathway.
... The CCDC family proteins contains coiled-coil structural domains that participates in various biologically functions such as cell cycle and regulation of gene expression. Changes in the structural domain of a CCDC gene or epigenetic changes are also detected in many tumors [60][61][62]. EPHX4 expression levels are highly upregulated in primary rectal cancer [63]. MMP1 is involved in the development of several cancers, and MMP1 upregulation promotes extracellular matrix degradation during the epithelial-mesenchymal transition and enhance migration and invasion of HCC cells [64]. ...
Article
Full-text available
Background & aims The effects of SARS-CoV-2 infection can be more complex and severe in patients with hepatocellular carcinoma (HCC) as compared to other cancers. This is due to several factors, including pre-existing conditions such as viral hepatitis and cirrhosis, which are commonly associated with HCC. Methods We conducted an analysis of epigenomics in SARS-CoV-2 infection and HCC patients, and identified common pathogenic mechanisms using weighted gene co-expression network analysis (WGCNA) and other analyses. Hub genes were identified and analyzed using LASSO regression. Additionally, drug candidates and their binding modes to key macromolecular targets of COVID-19 were identified using molecular docking. Results The epigenomic analysis of the relationship between SARS-CoV-2 infection and HCC patients revealed that the co-pathogenesis was closely linked to immune response, particularly T cell differentiation, regulation of T cell activation and monocyte differentiation. Further analysis indicated that CD4⁺ T cells and monocytes play essential roles in the immunoreaction triggered by both conditions. The expression levels of hub genes MYLK2, FAM83D, STC2, CCDC112, EPHX4 and MMP1 were strongly correlated with SARS-CoV-2 infection and the prognosis of HCC patients. In our study, mefloquine and thioridazine were identified as potential therapeutic agents for COVID-19 in combined with HCC. Conclusions In this research, we conducted an epigenomics analysis to identify common pathogenetic processes between SARS-CoV-2 infection and HCC patients, providing new insights into the pathogenesis and treatment of HCC patients infected with SARS-CoV-2.
... Through these domains, BRCA1 and BRCA2 form protein complexes involved in essential biological processes, such as the maintenance of the genomic integrity via homologous recombination (HR)-mediated DNA repair, the regulation of the oxidative stress and protein stability, and the modulation of gene transcription and cell cycle progression (7)(8)(9)(10)(11)(12)(13). Therefore, defective protein-protein interactions (PPIs) may lead to uncontrolled cell replication and genomic instability underlying BRCA1 and BRCA2 associated tumors (14,15). In particular, the PPI network of BRCA1 include the E2 ligase UbcH5a (MIM# 602961) which is engaged with BRCA1/BARD1 heterodimer (16) to preferentially catalyze atypical, non degradative K6-linked polyubiquitination related to DNA replication and repair (17)(18)(19)(20), and ABRAXAS (MIM# 611143) (21)(22)(23) which contributes to BRCA1-dependent DNA damage responses by mediating its recruitment to sites of DNA damage (22,24,25). Concerning BRCA2, DSS1 (MIM# 601285), a highly acidic 70 residue polypeptide, associates with a portion of the BRCA2 DBD encompassing the HD and the OB1 and OB2 motifs (6). ...
Article
Full-text available
The clinical screening of cancer predisposition genes has led to the identification of a large number of variants of uncertain significance (VUS). Multifactorial likelihood models that predict the odds ratio for VUS in favor or against cancer causality, have been developed, but their use is limited by the amount of necessary data, which are difficult to obtain for rare variants. The guidelines for variant interpretation of the American College of Medical Genetics and Genomics along with the Association for Molecular Pathology (ACMG/AMP) state that "well-established" functional studies provide strong support of a pathogenic or benign impact (criteria PS3 and BS3, respectively) and can be used as evidence type to reach a final classification. Moreover, the Clinical Genome Resource Sequence Variant Interpretation Working Group developed rule specifications to refine the PS3/BS3 criteria. Recently, Lira PC et al. developed the "Hi Set" approach that generated PS3/BS3 codes for over two-thousands BRCA1 VUS. While highly successful, this approach did not discriminate a group of variants with conflicting evidences. Here, we aimed to implement the outcomes of the "Hi-set" approach applying Green Fluorescent Protein (GFP)-reassembly assays, assessing the effect of variants in the RING and BRCT domains of BRCA1 on the binding of these domains with the UbcH5a or ABRAXAS proteins, respectively. The analyses of 26 clinically classified variants, including 13 tested in our previous study, showed 100% sensitivity and specificity in identifying pathogenic and benign variants for both the RING/UbcH5a and the BRCTs/ABRAXAS interactions. We derived the strength of evidences generated by the GFP-reassembly assays corresponding to moderate for both PS3 and BS3 criteria assessment. The GFP-reassembly assays were applied to the functional characterization of 8 discordant variants from the study by Lyra et al. The outcomes of these analyses, combined with those reported in the "Hi Set" study, allowed the assignment of ACMG/AMP criteria in favor or against pathogenicity for all 8 examined variants. The above findings were validated with a semi-quantitative Mammalian Two-Hybrid approach, and totally concordant results were observed. Our data contributes in shedding light on the functional significance of BRCA1 VUS and on their clinical interpretation within the ACMG/AMP framework.
Article
The maintenance of lysosome homeostasis is crucial for cell growth. Lysosome-dependent degradation and metabolism sustain tumor cell survival. Here, we demonstrate that CCDC50 serves as a lysophagy receptor, promoting tumor progression and invasion by controlling lysosomal integrity and renewal. CCDC50 monitors lysosomal damage, recognizes galectin-3 and K63-linked polyubiquitination on damaged lysosomes, and specifically targets them for autophagy-dependent degradation. CCDC50 deficiency causes the accumulation of ruptured lysosomes, impaired autophagic flux, and superfluous reactive oxygen species, consequently leading to cell death and tumor suppression. CCDC50 expression is associated with malignancy, progression to metastasis, and poor overall survival in human melanoma. Targeting CCDC50 suppresses tumor growth and lung metastasis, and enhances the effect of BRAFV600E inhibition. Thus, we demonstrate critical roles of CCDC50-mediated clearance of damaged lysosomes in supporting tumor growth, hereby identifying a potential therapeutic target of melanoma.
Article
CCDC (coiled-coil domain-containing) is a coiled helix domain that exists in natural proteins. There are about 180 CCDC family genes, encoding proteins that are involved in intercellular transmembrane signal transduction and genetic signal transcription, among other functions. Alterations in expression, mutation, and DNA promoter methylation of CCDC family genes have been shown to be associated with the pathogenesis of many diseases, including primary ciliary dyskinesia, infertility, and tumors. In recent studies, CCDC family genes have been found to be involved in regulation of growth, invasion, metastasis, chemosensitivity, and other biological behaviors of malignant tumor cells in various cancer types, including nasopharyngeal carcinoma, lung cancer, colorectal cancer, and thyroid cancer. In this review, we summarize the involvement of CCDC family genes in tumor pathogenesis and the relevant upstream and downstream molecular mechanisms. In addition, we summarize the potential of CCDC family genes as tumor therapy targets. The findings discussed here help us to further understand the role and the therapeutic applications of CCDC family genes in tumors.
Article
Mutations in breast cancer-associated 1 (BRCA1) increase the lifetime risk of developing breast cancer by up to 51% over the risk of the general population. Many aspects of this multifunctional protein have been revealed, including its essential role in homologous recombination repair, E3 ubiquitin ligase activity, transcriptional regulation, and apoptosis. Although most studies have focused on BRCA1 deficiency due to mutations, only a minority of patients carry BRCA1 mutations. A recent study has suggested an expanded definition of BRCA1 deficiency with reduced BRCA1 levels, which accounts for almost half of all triple-negative breast cancer (TNBC) patients. Reduced BRCA1 levels can result from epigenetic modifications or increased proteasomal degradation. In this review, we discuss how this knowledge of BRCA1 function and regulation of BRCA1 protein stability can help overcome the challenges encountered in the clinic and advance current treatment strategies for BRCA1-related breast cancer patients, especially focusing on TNBC.
Article
Full-text available
The Brca1 (breast cancer gene 1) tumor suppressor protein is phosphorylated in response to DNA damage. Results from this study indicate that the checkpoint protein kinase ATM (mutated in ataxia telangiectasia) was required for phosphorylation of Brca1 in response to ionizing radiation. ATM resides in a complex with Brca1 and phosphorylated Brca1 in vivo and in vitro in a region that contains clusters of serine-glutamine residues. Phosphorylation of this domain appears to be functionally important because a mutated Brca1 protein lacking two phosphorylation sites failed to rescue the radiation hypersensitivity of a Brca1-deficient cell line. Thus, phosphorylation of Brca1 by the checkpoint kinase ATM may be critical for proper responses to DNA double-strand breaks and may provide a molecular explanation for the role of ATM in breast cancer.
Article
Full-text available
Chk1, an evolutionarily conserved protein kinase, has been implicated in cell cycle checkpoint control in lower eukaryotes. By gene disruption, we show that CHK1 deficiency results in a severe proliferation defect and death in embryonic stem (ES) cells, and peri-implantation embryonic lethality in mice. Through analysis of a conditional CHK1-deficient cell line, we demonstrate that ES cells lacking Chk1 have a defective G(2)/M DNA damage checkpoint in response to gamma-irradiation (IR). CHK1 heterozygosity modestly enhances the tumorigenesis phenotype of WNT-1 transgenic mice. We show that in human cells, Chk1 is phosphorylated on serine 345 (S345) in response to UV, IR, and hydroxyurea (HU). Overexpression of wild-type Atr enhances, whereas overexpression of the kinase-defective mutant Atr inhibits S345 phosphorylation of Chk1 induced by UV treatment. Taken together, these data indicate that Chk1 plays an essential role in the mammalian DNA damage checkpoint, embryonic development, and tumor suppression, and that Atr regulates Chk1.
Article
Full-text available
The recent discovery of checkpoint kinases has suggested the conservation of checkpoint mechanisms between yeast and mammals. In yeast, the protein kinase Chk1 is thought to mediate signaling associated with the DNA damage checkpoint of the cell cycle. However, the function of Chk1 in mammals has remained unknown. Targeted disruption of Chk1 in mice showed that Chk1(-/-) embryos exhibit gross morphologic abnormalities in nuclei as early as the blastocyst stage. In culture, Chk1(-/-) blastocysts showed a severe defect in outgrowth of the inner cell mass and died of apoptosis. DNA replication block and DNA damage failed to arrest the cell cycle before initiation of mitosis in Chk1(-/-) embryos. These results may indicate that Chk1 is indispensable for cell proliferation and survival through maintaining the G(2) checkpoint in mammals.
Article
Full-text available
Hereditary breast and ovarian cancer syndromes can be caused by loss-of-function germline mutations in one of two tumour-suppressor genes, BRCA1 and BRCA2 (ref. 1). Each gene product interacts with recombination/DNA repair proteins in pathways that participate in preserving intact chromosome structure. However, it is unclear to what extent such functions specifically suppress breast and ovarian cancer. Here we analyse what is known of BRCA gene function and highlight some unanswered questions in the field.
Article
Full-text available
Cell cycle arrests in the G1, S, and G2phases occur in mammalian cells after ionizing irradiation and appear to protect cells from permanent genetic damage and transformation. Though Brca1 clearly participates in cellular responses to ionizing radiation (IR), conflicting conclusions have been drawn about whether Brca1 plays a direct role in cell cycle checkpoints. Normal Nbs1 function is required for the IR-induced S-phase checkpoint, but whether Nbs1 has a definitive role in the G2/M checkpoint has not been established. Here we show that Atm and Brca1 are required for both the S-phase and G2 arrests induced by ionizing irradiation while Nbs1 is required only for the S-phase arrest. We also found that mutation of serine 1423 in Brca1, a target for phosphorylation by Atm, abolished the ability of Brca1 to mediate the G2/M checkpoint but did not affect its S-phase function. These results clarify the checkpoint roles for each of these three gene products, demonstrate that control of cell cycle arrests must now be included among the important functions of Brca1 in cellular responses to DNA damage, and suggest that Atm phosphorylation of Brca1 is required for the G2/M checkpoint.
Article
Full-text available
We generated mouse mutants carrying in the Brca1 locus a modification (Brca1(tr)) that eliminates the C-terminal half of the protein product and obtained results indicating that, depending on genetic background, the missing BRCT and/or other domains are dispensable for survival, but essential for tumor suppression. Most of the apparently hypomorphic Brca1(tr/tr) mutants developed various tumors. Lymphomas were detected at all ages, whereas sarcomas and carcinomas, including breast cancer, appeared after a long latency. The mammary tumors showed striking variability in histopathological patterns suggesting stochastic engagement of tumorigenic pathways in their progression, to which the Brca1(tr/tr) mutation was apparently a late participant.
Article
The bph locus of Pseudomonas sp. LB400, encoding biphenyl/polychlorinated biphenyl (PCB) degradation, contains a region of about 3.5 kb of hitherto unknown function, between bphC and bphD. This DNA segment has now been characterized. Four structural genes have been located and identified by a combination of expression cloning, enzyme activity tests and DNA sequencing. The region contains four closely spaced cistrons (bphKHJI) encoding a glutathione S-transferase (GST), a 2-hydroxypenta-2,4-dienoate hydratase, an acetaldehyde dehydrogenase (acylating) and a 4-hydroxy-2-oxovalerate aldolase, respectively. The latter three are enzymes required for conversion of the aliphatic end product of bphABCD-encoded catabolism of biphenyls to Krebs cycle intermediates. The discovery of these genes provides a rationale for growth of the strain on chlorinated biphenyls which yield chlorinated benzoates as dead-end metabolites. The sequences of the enzymes involved are 54-71% identical to those of homologous enzymes encoded by the dmp and xyl operons. The role of the GST in the degradation of biphenyls is less clear, but since it was found to contain, in the putative xenobiotic substrate-binding domain, a region which shares about 29% of identical amino acids with a bacterial tetrachlorohydroquinone dehalogenase, it may be involved in dehalogenation of PCB-degradative intermediates.
Article
Germline mutations of BRCA1 predispose women to breast and ovarian cancers. BRCA1 contains several functional domains that interact directly or indirectly with a variety of molecules, including tumor suppressors (p53, RB, BRCA2 and ATM), oncogenes (c-Myc, casein kinase II and E2F), DNA damage repair proteins (RAD50 and RAD51), cell-cycle regulators (cyclins and cyclin-dependent kinases), transcriptional activators and repressors (RNA polymerase II, RHA, histone deacetylase complex and CtIP) and others. Mounting evidence indicates that these physical associations are not artifacts; rather, BRCA1 is likely to serve as an important central component in multiple biological pathways that regulate cell-cycle progression, centrosome duplication, DNA damage repair, cell growth and apoptosis, and transcriptional activation and repression. This review examines our understanding of the significance of the interactions between BRCA1 and other proteins, through which BRCA1 maintains genome integrity and represses tumor formation. Published 2000 John Wiley & Sons, Inc.
Article
The inability to repair DNA damage properly in mammals leads to various disorders and enhanced rates of tumour development. Organisms respond to chromosomal insults by activating a complex damage response pathway. This pathway regulates known responses such as cell-cycle arrest and apoptosis (programmed cell death), and has recently been shown to control additional processes including direct activation of DNA repair networks.
Article
BRCA1 interacts in vivo with a novel protein, BACH1, a member of the DEAH helicase family. BACH1 binds directly to the BRCT repeats of BRCA1. A BACH1 derivative, bearing a mutation in a residue that was essential for catalytic function in other helicases, interfered with normal double-strand break repair in a manner that was dependent on its BRCA1 binding function. Thus, BACH1/BRCA1 complex formation contributes to a key BRCA1 activity. In addition, germline BACH1 mutations affecting the helicase domain were detected in two early-onset breast cancer patients and not in 200 matched controls. Thus, it is conceivable that, like BRCA1, BACH1 is a target of germline cancer-inducing mutations.