ArticlePDF Available

CECR2 Is Involved in Spermatogenesis and Forms a Complex with SNF2H in the Testis

Authors:

Abstract and Figures

The regulation of nucleosome positioning and composition by ATP-dependent chromatin remodeling enzymes and their associated binding partners plays important biological roles in mammals. CECR2 is a binding partner to the ISWI (imitation switch) ATPase SNF2L/SMARCA1 and is involved in neural tube closure and inner ear development; however, its functions in adult tissues have not been examined. Here, we report that CECR2 contributes to spermatogenesis and forms a complex that includes the other ISWI ATPase SNF2H/SMARCA5 in the testis. Cecr2 mutant males non-penetrant for neural tube defects sired smaller litters than wild-type males. Strikingly, while we found that Cecr2 mutants have normal seminiferous epithelium morphology, sperm count, motility, and morphology, the mutant spermatozoa were compromised in their ability to fertilize oocytes. Investigation of CECR2/ISWI complexes in the testis showed that SNF2H interacted with CECR2, and this interaction was also observed in embryonic stem cells, suggesting that CECR2 may interact with SNF2H or SNF2L depending on the cell type. Finally, we found that Cecr2 mutants exhibit misregulation of the homeobox transcription factor Dlx5 in the testis, suggesting that CECR2 complexes may regulate gene expression during spermatogenesis. Taken together, our results demonstrate a novel role of CECR2-containing complexes in spermatogenesis and show that CECR2 interacts predominantly with SNF2H instead of SNF2L in the testis.
Content may be subject to copyright.
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elsevier’s archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/copyright
Author's personal copy
CECR2 Is Involved in Spermatogenesis and Forms a
Complex with SNF2H in the Testis
Peter J. Thompson, Kacie A. Norton, Farshad H. Niri,
Christine E. Dawe and Heather E. McDermid
Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2E9
Received 14 March 2011;
received in revised form
22 November 2011;
accepted 22 November 2011
Available online
2 December 2011
Edited by J. Karn
Keywords:
CECR2;
SNF2H/SMARCA5 SNF2L/
SMARCA1;
ISWI;
spermatogenesis;
chromatin remodeling
The regulation of nucleosome positioning and composition by ATP-
dependent chromatin remodeling enzymes and their associated binding
partners plays important biological roles in mammals. CECR2 is a
binding partner to the ISWI (imitation switch) ATPase SNF2L/SMARCA1
and is involved in neural tube closure and inner ear development;
however, its functions in adult tissues have not been examined. Here, we
report that CECR2 contributes to spermatogenesis and forms a complex
that includes the other ISWI ATPase SNF2H/SMARCA5 in the testis.
Cecr2 mutant males non-penetrant for neural tube defects sired smaller
litters than wild-type males. Strikingly, while we found that Cecr2
mutants have normal seminiferous epithelium morphology, sperm count,
motility, and morphology, the mutant spermatozoa were compromised in
their ability to fertilize oocytes. Investigation of CECR2/ISWI complexes
in the testis showed that SNF2H interacted with CECR2, and this
interaction was also observed in embryonic stem cells, suggesting that
CECR2 may interact with SNF2H or SNF2L depending on the cell type.
Finally, we found that Cecr2 mutants exhibit misregulation of the
homeobox transcription factor Dlx5 in the testis, suggesting that CECR2
complexes may regulate gene expression during spermatogenesis. Taken
together, our results demonstrate a novel role of CECR2-containing
complexes in spermatogenesis and show that CECR2 interacts predom-
inantly with SNF2H instead of SNF2L in the testis.
© 2011 Elsevier Ltd. All rights reserved.
Introduction
A key mechanism controlling the composition,
dynamics, and positioning of nucleosomes in eukary-
otes is the activity of ATP-dependent chromatin
remodeling complexes.
1
The ISWI (imitation switch)
family of ATPases consists of two homologues in
mammals, SNF2H/SMARCA5 and SNF2L/
SMARCA1, which exhibit 80% identity at the
protein level.
2
There are a total of eight known
mammalian ISWI complexes identified in human
and/or mouse cells, in which either SNF2H or SNF2L
is incorporated as the catalytic subunit. SNF2H is in
six of these complexes (ACF, CHRAC, WICH, WCRF,
RSF, and NoRC
38
), while SNF2L is in NURF and
CERF.
9,10
In vivo, ISWI complexes have many diverse
functions including replication of pericentric hetero-
chromatin,
11,12
ribosomal RNA gene repression,
13
developmental gene regulation,
9,14
and maintenance
of chromosome structure.
15
Genetic studies in mice
suggest that SNF2H-containing complexes are
*Corresponding author. University of Alberta, G-508
Biological Sciences Building, Edmonton, Alberta, Canada
T6G 2E9. E-mail address: heather.mcdermid@ualberta.ca.
Present address: P. J. Thompson, Department of
Medical Genetics, The University of British Columbia,
Vancouver, British Columbia, Canada V6T 1Z3.
Abbreviations used: Xgal, 5-bromo-4-chloro-3-
indolyl-β,D-galactopyranoside; RT, reverse transcriptase;
qRT-PCR, quantitative RT-PCR; IP, immunoprecipitation.
doi:10.1016/j.jmb.2011.11.041 J. Mol. Biol. (2012) 415, 793806
Contents lists available at www.sciencedirect.com
Journal of Molecular Biology
journal homepage: http://ees.elsevier.com.jmb
0022-2836/$ - see front matter © 2011 Elsevier Ltd. All rights reserved.
Author's personal copy
essential for early development,
16
while SNF2L-
containing complexes are involved in oogenesis.
17
The SNF2L-containing complex CERF was puri-
fied from human embryonic kidney cells (HEK293)
and includes the bromodomain-containing protein
CECR2.
10
CECR2-containing complexes play im-
portant roles in embryogenesis, since mutations in
Cecr2 causes neural tube and inner ear defects;
10,18,19
however, the role of CECR2 in adult tissues has not
been investigated. In the present study, we investi-
gated the role of CECR2 in the male germ line and
report that CECR2 is involved in spermatogenesis.
In mutant males without neural tube defects, Cecr2
was highly expressed in adult spermatogonia, and
Cecr2 mutant males sired significantly smaller litters
than wild-type females. Loss of Cecr2 in males
resulted in a lower percentage of fertilized oocytes,
although those successfully fertilized appeared to
develop to blastocysts in culture. We also found that
CECR2 interacted with SNF2H, rather than SNF2L,
in the adult testis. We observed a similar interaction
with SNF2H in embryonic stem cells, raising the
possibility that CECR2 incorporates into complexes
with either SNF2H or SNF2L depending on the cell
type. Finally, we found that loss of functional
CECR2 affects expression of the homeobox tran-
scription factor Dlx5 in the testis. Together, our
results indicate that CECR2 contributes to sper-
matogenesis and forms complexes with SNF2H in
the testis.
Results
Mutation of Cecr2 affects litter sizes but does
not cause gross morphological changes to the
seminiferous epithelium or mature spermatozoa
BALB/c Cecr2
Gt45Bic
(Cecr2
Gt
) homozygous mu-
tants develop neural tube defects at 74% pene-
trance.
10
The 26% non-penetrant animals are
viable, but we noticed that some mutant males
were unable to sire pups. Two homozygous
mutant studs unable to sire pups were necropsied
and found to have compromised spermatogenesis
with an abnormally low cellular density in the
seminiferous epithelium, reduced spermatozoa per
tubule, and a lower than usual number of active
tubules (data not shown). Furthermore, there was a
notable reduction in the number of spermatozoa in
the epididymis and a large number of degenerate
seminiferous cells and cellular fragments mixed in
with the spermatozoa, suggesting a possible defect
in sperm development and maturation. This led us
to conduct a more comprehensive study of sper-
matogenesis and fertility in Cecr2
Gt
mutant males,
which revealed a more subtle phenotype in most
animals.
To examine whether Cecr2 is expressed in the male
germ line, we profiled the expression of the mutant
reporter protein CECR2
Gt
during testis develop-
ment in homozygous mutant embryos and adults
non-penetrant for neural tube defects. CECR2
Gt
is
the CECR2β-galactosidase fusion protein product
of Cecr2
Gt
in which the 290 N-terminal amino acids
encoding the DDT domain and AT-hook of CECR2
are spliced to a βgeo cassette, effectively deleting the
bromodomain and C-terminal two-thirds of the
protein (Fig. 1a). 5-Bromo-4-chloro-3-indolyl-β,D-
galactopyranoside (Xgal) staining revealed Cecr2
expression as early as E16.5 in the gonocytes of the
sex cords (Fig. 1b). Expression persisted in the
gonocytes throughout the development of the testes
and remained high in the adult, where staining was
strong in spermatogonia, lower in spermatocytes
and was not present in further differentiated cells
such as the elongate spermatids (Fig. 1b). While
there was no CECR2
Gt
staining in the Leydig cells at
E16.5, staining was evident in Leydig cells of the
adult testis (data not shown). Wild-type males
showed no endogenous staining in the testes
(Fig. 1b). To assess whether the Cecr2
Gt
mutants
exhibited any abnormal phenotypes related to
spermatogenesis and fertility, we determined litter
sizes produced by Cecr2
Gt
homozygous mutants
compared to wild type when each was mated to
wild-type females (Fig. 1c). We found that, on
average, mutants sired significantly smaller litters
(average of 4.5 pups in 22 litters) compared to wild
type (average of 6.5 pups in 70 litters) (Fig. 1c;
p=0.022). The range of litter sizes produced by wild-
type males approximated a normal distribution as
expected. In contrast, five out of the eleven Cecr2
Gt
non-penetrant males tested (45.5%) never produced
a litter greater than four pups, and one male out of
the eleven males tested was unable to sire any pups
during the test period (two females were tested). All
23 wild-type males tested were able to produce
litters greater than four at least once in their
breeding history, and no sterile matings were
observed. The length of time for Cecr2
Gt
mutant
males to successfully mate with a female was
generally within 14 days and was comparable to
that of wild-type individuals within our colony.
Mating behavior in the mutants appeared similar to
that of wild-type males (data not shown). Taken
together, these data suggest that CECR2 contributes
to fertility.
We next ascertained whether the sub-fertility
phenotype of Cecr2 mutants could be explained by
abnormal spermatogenesis, such as a defect in either
self-renewal or meiotic differentiation of spermato-
gonia, where CECR2 was strongly expressed
(Fig. 1b). We calculated the spermatidspermatogo-
nia (SDSG) ratio in five of the fertility-tested Cecr2
mutants compared to four wild-type males, since
this ratio would indicate defects in spermatogonia
794 CECR2 Is Involved in Spermatogenesis
Author's personal copy
self-renewal as a larger SDSG ratio compared to
wild type or defects in differentiation as smaller SD
SG ratio compared to wild type. However, we found
that there was no significant difference in the SDSG
ratios (Fig. 2a; p= 0.102), and both wild-type and
mutant males had ratios of 2, consistent with
normal spermatogonia self-renewal and differentia-
tion. In addition, examination of the seminiferous
epithelium of Cecr2 mutants did not reveal any obvi-
ous abnormalities in cellular morphology compared
Fig. 1. Cecr2
Gt
mutants sire significantly smaller litters than wild-type males. (a) Schematic of wild-type CECR2 protein
compared to the CECR2
Gt
mutant reporter protein. DDT, domain in different transcription factors; AT-hook, AT-rich
sequence binding motif; NLS, nuclear localization signal. (b) Xgal staining pattern of CECR2
Gt
in the male gonad at E16.5
and E19.5, at postnatal day 0 (P0), and in adult (6 weeks old). Black arrows indicate CECR2
Gt
staining in gonocytes
(E19.5 and P0) and spermatogonia (adult), respectively. Wild-type tissues (Cecr2
+/+
) were Xgal stained as negative
controls. (c) Histogram displays the average number of pups per litter sired by either BALB/c wild-type males (70 litters)
or Cecr2
Gt
mutant males (22 litters) where both were crossed with wild-type females. Frequency plots show the same data
distributed by frequency of litter size for wild type (upper plot) or Cecr2 mutants (lower plot). Error bars represent SD,
and asterisks indicate p=0.022, two-tailed T-test.
795CECR2 Is Involved in Spermatogenesis
Author's personal copy
to wild type (Fig. 2b). Furthermore, we assessed
whether changes in spermatozoa numbers or motil-
ity could underlie the reduced fertility in Cecr2
mutants. However, spermatozoa collected from the
cauda epididymis of Cecr2 mutants appeared
morphologically normal and motile when compared
to wild-type males (data not shown). From these
data, we conclude that CECR2 contributes to fertility
in a manner that usually does not drastically alter
spermatogonia self-renewal or differentiation. Thus,
the original two infertile males examined were
atypical.
Cecr2 mutant spermatozoa are compromised in
their ability to fertilize oocytes
Since most Cecr2 mutants exhibited reduced
fertility without showing obvious defects in semi-
niferous tubule morphology, ratios of spermatids-
to-spermatogonia, or spermatozoa motility, we
asked whether the reduced fertility could be
explained by defects in oocyte fertilization or
compromised viability of embryos. We focused
our analysis on four mutant males and three
matched wild-type males, none of which were
included in the previous analysis. Fertility testing
of wild-type or mutant males with wild-type
females was performed by scoring embryos this
time rather than live births to determine whether the
reduction in litter size was due to an increase in
resorbed embryos or early death. Analysis con-
firmed that the mutants had a significant reduction
in the average number of live embryos compared to
wild type, which was comparable to our previous
analysis (Fig. 1c). Due to considerable inter-litter size
variation for each male, the litter sizes were pooled
for each genotype and compared by the Wilcoxon
Rank Sum test and found to be significantly
different (p= 0.0011; Table 1). Average litter size for
each male ranged from 6.00 to 6.88 for wild-type
males and from 2.86 to 5.29 for mutants. However,
we observed similar numbers of resorbed or dead
embryos in litters sired by wild-type and Cecr2
mutant males (Table 1). This suggests that increased
embryo death does not account for the decreased
litter sizes in mutant males.
We performed detailed analyses of sperm count,
morphology, and motility of mature sperm from the
cauda epididymis in the three wild-type males and
the three mutant males with the smallest average
litter size (Table 2). We did not observe any
significant differences in total sperm counts, motility
patterns, or morphology of mutant sperm compared
to wild type (Table 2). Therefore, loss of Cecr2 does
not result in visibly abnormal sperm.
Prior to sperm analysis, we used two wild-type
fertility-tested males and two mutant fertility-tested
males to determine whether preimplantation em-
bryos sired by Cecr2 mutants exhibited reduced
survival to the blastocyst stage or whether mutant
sperm exhibited a reduced ability to fertilize
oocytes. Wild-type FVB/N females were induced
to superovulate and then mated with either a wild-
type or a mutant male. The following day, oocytes
Fig. 2. Cecr2
Gt
mutants do not
have significant changes in sperma-
tidspermatogonia ratios or semi-
niferous epithelium morphology
compared to wild type. (a) Elongat-
ed spermatids and spermatogonia
from four BALB/c wild-type males
and five Cecr2
Gt
mutant males were
counted (blind to genotype), and a
ratio was calculated from images
(n=10) of seminiferous tubule
cross-section images for each male.
Each wild-type and Cecr2 mutant
male had been previously been
housed with at least one female.
Error bars are standard errors of the
mean (p=0.102, two-tailed T-test).
(b) Wild-type and Cecr2
Gtc
mutant
testes were sectioned and stained
with hematoxylin and eosin. Im-
ages show the seminiferous epithe-
lium at 630× magnification. Scale
bars represent 50 μm.
796 CECR2 Is Involved in Spermatogenesis
Author's personal copy
were isolated and cultured in vitro. Freshly isolated
oocytes were first observed under phase-contrast
microscopy to identify the number of zygotes that
had formed as evidenced by maternal and paternal
pronuclei. Zygotes were cultured for an additional
34 days, and the number of zygotes that survived
to blastocysts was recorded. While zygotes sired by
wild-type males showed 81% average survival to
blastocysts, zygotes sired by Cecr2 mutants had an
average survival of only 39% (pb0.0001; Table 3).
More importantly, however, we observed that the
number of fertilized oocytes was significantly
smaller for Cecr2 mutants compared to wild type
(40% versus 78%, respectively, and p= 0.0031;
Table 3). Furthermore, the survival of fertilized
oocytes to blastocysts was not different between
wild-type and Cecr2 mutant males (100% versus
81%, respectively; Table 3). These data indicate
that mutation of Cecr2 compromises the ability of
sperm to fertilize oocytes and that reduced fertili-
zation underlies the observed reduction of surviving
blastocysts sired by Cecr2 mutants.
Analysis of the mutant protein CECR2
Gt
reveals
different isoforms in ES cells compared to adult
testis
We next investigated the composition of endoge-
nous CECR2-containing complexes in the germ line.
Since our previous attempts in generating polyclon-
al antibodies to mouse CECR2 were unsuccessful,
we chose to investigate the CECR2
Gt
reporter
protein instead (Fig. 1a). Importantly, CECR2
Gt
retains the DDT domain, which is necessary for
the interaction between ISWI proteins and their
binding partners, as shown for Drosophila ACF1 and
ISWI
20
and human RSF1 and SNF2H.
21
CECR2
Gt
also retains the AT-hook, which typically functions
as an AT-rich DNA-binding domain.
22
Further-
more, although CECR2
GT
lacks the putative nuclear
localization signal of CECR2, immunofluorescence
analysis on the Cecr2
Gt45Bic
heterozygous mouse ES
cell line CT45 showed that it is retained in the nucleus
and co-localizes with mitotic chromosomes.
23
Based
on these observations, we determined that CECR2
Gt
would be a suitable endogenous reporter protein for
investigating CECR2/ISWI complexes in vivo.
We performed Western blot analysis using β-
galactosidase antibodies on nuclear extracts pre-
pared from CT45 ES cells, E13.5 embryos, perinatal
brain, and a panel of adult tissues (Fig. 3a and b).
CECR2
Gt
was specifically detected as a single band
at the expected size of 180 kDa in whole embryos
and adult testis (Fig. 3a), but levels were low or
undetectable in perinatal brain, adult brain, liver,
and kidney. In CT45 cells, CECR2
Gt
was present as a
faster migrating band at 160 kDa (Fig. 3b). Since
the differences in apparent protein size could be due
to alternative splicing within the first seven exons of
Table 1. Fertility analysis of wild-type and Cecr2 mutant males
Cecr2
+/+
506 Cecr2
+/+
2019 Cecr2
+/+
2020 Cecr2
Gt/Gt
1934 Cecr2
Gt/Gt
2018 Cecr2
Gt/Gt
2056 Cecr2
Gt/Gt
2058
Embryos Resorbed Embryos Resorbed Embryos Resorbed Embryos Resorbed Embryos Resorbed Embryos Resorbed Embryos Resorbed
60100 145021 31 20
63 70 814120 61 12
70 901002330 70 24
70 22 805230 20 30
53 62 526131 12110
81 14 703221110 20
81 71 704250 70100
61 9062
40 71
80
6.50 0.90 6.00 1.29 6.88 0.88 4.67 1.56 2.86 0.43 5.29 0.57 4.43 0.86
Litter sizes were averaged for each genotype to assess statistical significance
Wild type Mutant Significance
Average litter size 6.46 4.31 p=0.0011
Average resorbed 1.02 0.85 Not significant
797CECR2 Is Involved in Spermatogenesis
Author's personal copy
Table 3. Fertilization and blastocyst survival of embryos sired by wild-type and Cecr2 mutant males
Genotype Pronuclei Blastocysts Total oocytes Pronuclei/total (%) Blastocysts/total (%) Blastocysts/pronuclei (%)
Cecr2
+/+
506 7 7 8 87.5 87.5 100.0
2 3 5 40.0 60.0 100.0
7 7 9 77.8 77.8 100.0
13 15 17 76.5 88.2 100.0
10 17 58.8
12 14 85.7
13 23 56.5
16 16 100.0
78 87.5
4 4 4 100.0 100.0 100.0
Cecr2
+/+
2020 9 12 21 42.9 57.1 100.0
14 14 14 100.0 100.0 100.0
13 14 92.9
12 13 92.3
916 56.3
6 6 6 100.0 100.0 100.0
Average Cecr2
+/+
10 11 14 78.1
a
81.3
b
100.0
c
Cecr2
Gt/Gt
1934 0 2 18 0.0 11.1 100.0
5 4 9 55.6 44.4 79.9
6 8 13 46.2 61.5 100.0
620 30.0
621 28.6
14 18 77.8
823 34.8
2 1 5 40.0 20.0 50.0
6 0 10 60.0 0.0 0.0
Cecr2
Gt/Gt
2018 6 10 20 30.0 50.0 100.0
5 5 8 62.5 62.5 100.0
810 80.0
11 17 64.7
11 19 57.9
10 20 50.0
3 4 10 30.0 40.0 100.0
4 6 10 40.0 60.0 100.0
Average Cecr2
Gt/Gt
4 7 15 40.0
a
38.8
b
81.1
c
c
pN0.05, MannWhitney test.
Bold highlights the average values across the rows.
, pronuclei not scored.
approximates 100%, number of blastocysts slightly exceeded pronuclei.
a
pb0.001, MannWhitney test.
b
p=0.0031, MannWhitney test.
Table 2. Analysis of mature sperm of wild-type and Cecr2 mutant males
Genotype
Cecr2
+/+
506
Cecr2
+/+
2019
Cecr2
+/+
2020
Cecr2
Gt/Gt
1934
Cecr2
Gt/Gt
2018
Cecr2
Gt/Gt
2058
Average
Cecr2
+/+
Average
Cecr2
Gt/Gt
T-test
(p-value)
Sperm count
10
6
cells/ml)
7.6 9.5 6.7 6.3 6.0 7.0 7.9 6.4 0.162
SD 0.8 0.6 0.9 1.1 0.4 0.4 1.4 0.5
Motility (%)
Moving forward 53.2 47.1 50.2 64.0 37.6 45.4 50.2 49.0 0.891
Moving but
not forward
3.6 7.8 3.6 3.0 6.3 5.4 5.0 4.9 0.956
Tail motion only 39.2 29.9 35.9 24.5 44.9 39.0 35.0 36.1 0.873
Not moving 4.1 15.2 10.3 8.5 11.2 10.2 9.9 10.0 0.977
Morphology (%)
Linear/normal 67.6 76.1 70.8 69.2 75.5 61.3 71.5 68.7 0.587
Angulated
(N90°/hairpin)
21.7 13.7 17.2 20.7 16.5 25.0 17.5 20.7 0.397
Headless 10.7 10.2 12.0 10.1 8.0 13.7 11.0 10.6 0.844
SD=standard deviation.
798 CECR2 Is Involved in Spermatogenesis
Author's personal copy
Cecr2
Gt45Bic
transcripts, we performed reverse tran-
scriptase (RT)-PCR analysis of exons 17from
heterozygous adult testis and CT45 cells (Fig. 3c).
In both CT45 cells and testis, exons 17 were present
in Cecr2
Gt45Bic
transcripts, suggesting that the
differences in protein size may be due to posttrans-
lational modifications of CECR2
Gt
and, by exten-
sion, CECR2.
CECR2
Gt
forms distinct complexes with SNF2H
in the adult testis and ES cells
Human CECR2 interacts with SNF2L in embryonic
kidney cells;
10
therefore, we determined whether
mouse CECR2 also forms a complex with SNF2L or
whether it interacts with the other ISWI homologue
SNF2H in the testis. We used an immunoaffinity
approach to enrich for CECR2
Gt
-associated proteins
from testis nuclear extracts prepared from Cecr2
Gt45Bic
heterozygotes (Fig. 4a). Nuclear proteins were
first passed over a protein G-Sepharose column,
containing no antibody (mock), and then over a
β-galactosidase antibody column. The columns
were washed with a high salt buffer (containing
0.4 M NaCl) to minimize nonspecific binding,
and elution was performed at low pH. Western
blot analysis of the eluted fractions from each
column showed that the column retained CECR2
Gt
and SNF2H but not SNF2L (Fig. 4a). Consistent
with the idea that CECR2
Gt
interacts with SNF2H
independently of other SNF2H-associated proteins,
the immunoaffinity elutions did not contain detect-
able levels of ACF1 from ACF and CHRAC, WSTF
from WICH, RSF1 from RSF, or TIP5 from NoRC
(Fig. 4a). Similarly, TATA-binding protein was
undetectable in the elutions, consistent with
previous findings showing that ISWI complexes
including CERF do not contain general transcription
factors.
37,9,10
These data indicate that CECR2
Gt
interacts with SNF2H rather than SNF2L in the
adult testis and that the CECR2
Gt
/SNF2H
complex is independent of other SNF2H-containing
complexes.
We next assessed the native molecular weight of
the CECR2
Gt
/SNF2H complex by fractionating
heterozygous testis nuclear extracts over a gel-
filtration column equilibrated in a high salt buffer
(Fig. 4b). Western blot analysis of the fractions
showed that CECR2
Gt
eluted with the peak centered
at 0.91 MDa and was not detectable in the
fractions corresponding to the monomeric size
(200 kDa, fractions 4244), suggesting that almost
all of it was incorporated into this complex (Fig. 4b).
SNF2H partially co-eluted with CECR2
Gt
, and the
peak of SNF2H eluted at a molecular mass of
600700 kDa, consistent with the sizes of other
SNF2H-containing complexes such as ACF,
CHRAC, WICH, and NoRC.
58
Immunoprecipita-
tion (IP) of SNF2H from the co-eluting fractions
also precipitated CECR2
Gt
, demonstrating that these
Fig. 3. Analysis of the mutant CECR2
Gt
reporter protein. (a) Western blot of CECR2
Gt
in nuclear extracts (40 μg per
lane) prepared from E13.5 wild-type (+/+) and Cecr2
Gt
homozygous mutant embryos and adult testes. Molecular mass
markers (kDa) are shown to the left of the blots. (b) Western blot as in (a) except on nuclear extracts prepared from CT45
ES cells, three E13.5 Cecr2
Gt
heterozygous embryos, and Cecr2
Gt
heterozygous postnatal brain (P0), adult brain, liver,
kidney, and testis. Asterisk indicates a nonspecific band. (c) RT-PCR analysis of exons 17inCecr2
Gt45Bic
transcripts from
CT45 ES cells and heterozygous adult testis. Negative controls were performed by omitting RT from the cDNA synthesis
reaction. The amplicon size is 817 bp, and identification of the band as Cecr2 exons 17 was confirmed by sequencing.
RT, no RT negative controls; M, DNA fragment size marker.
799CECR2 Is Involved in Spermatogenesis
Author's personal copy
fractions contain the CECR2
Gt
/SNF2H complex
(Fig. 4b). Treatment of the extract with ethidium
bromide did not alter the incorporation of CECR2
Gt
into this high-molecular-weight complex (Fig. 4c),
suggesting that the complex assembles independent-
ly of DNA. In addition, the high-molecular-weight
Fig. 4 (legend on next page)
800 CECR2 Is Involved in Spermatogenesis
Author's personal copy
CECR2
Gt
-containing complex appeared to be inde-
pendent of wild-type CECR2, since CECR2
Gt
eluted
around the same molecular weight in Cecr2 homo-
zygous mutant testis nuclear extract (Fig. 4d).
Finally, the other SNF2H-containing complexes
appeared unperturbed in the Cecr2 mutant testis
nuclear extracts, as the elution profile of SNF2H was
very similar compared to that of the heterozygous
testis with the peak elution fractions at 600
700 kDa (Fig. 4d).
Finally, we chose to examine whether a similar
CECR2/SNF2H-containing complex is present in ES
cells, since we found that CECR2
Gt
was a different
size in ES cells compared to adult testes and whole
embryos (Fig. 3b). Fractionation of the CT45 ES cell
nuclear extract showed that CECR2
Gt
eluted over a
broad range of sizes from 200 to 800 kDa (fractions
3642), with the peak centered at 300400 kDa in
fraction 40, which was similar to the range of sizes
where SNF2H eluted (Fig. 4e). In contrast to adult
testis, a relatively large amount of CECR2
Gt
was
detected in fractions corresponding to the mono-
meric size at 67200 kDa in fractions 4246,
suggesting that not all the CECR2
Gt
is incorporated
into complexes in ES cells. IP of SNF2H from the
CECR2
Gt
peak fractions at the size range of 300
400 kDa also pulled down CECR2
Gt
, demonstrating
that these fractions contain the complex (Fig. 4e).
Taken together, these results suggest that CECR2
Gt
forms distinct complexes with SNF2H in both the
adult testis and ES cells.
Mutation of Cecr2 results in reduced expression
of Dlx5
To investigate the cause of the fertilization defect
in Cecr2
Gt
mutant spermatozoa, we hypothesized
that there may be misregulation of genes encoding
factors involved in fertility. To determine a set of
candidate genes to test, we took advantage of our
previously generated microarray data set from E8.5
wild-type and Cecr2
Gt
mutant embryos.
18
Although
these embryos were female and prior to gonad
formation, we reasoned that there may be genes that
Fig. 4. CECR2
Gt
interacts in complexes with SNF2H in adult testes and ES cells. (a) Immunoaffinity enrichment of
CECR2
Gt
and associated proteins from Cecr2
Gt
heterozygous adult testis nuclear extract. Eluted fractions were analyzed
by Western blot to detect the indicated proteins. Loadrepresents 0.6% of material loaded onto the column.
Approximately 7% of each fraction was loaded in the lanes. (b) Upper: gel-filtration fractionation and Western blot of
CECR2
Gt
and SNF2H in Cecr2
Gt
heterozygous testis nuclear extract. INis 1% of material loaded onto the column,
V
O
is the void volume, and fraction numbers are indicated above the blot, with pooled fractions shown in boldface. The
CECR2
Gt
peak fraction 32 corresponded to a size of 0.91 MDa. Lower: IP of SNF2H from the CECR2
Gt
peak fractions
3133, followed by Western blot. IgGis the purified mouse IgG negative control IP. (c) Gel filtration and Western blot of
CECR2
Gt
as in (b) except that ethidium bromide was added to the testis nuclear extract before gel filtration. (d) Gel
filtration and Western blot as in (b) except using Cecr2
Gt
homozygous mutant testis nuclear extract. (e) Upper: gel
filtration and Western blot as in (b) except using CT45 ES cell nuclear extract. The CECR2
Gt
peak fraction 40 corresponded
to a size of 300400 kDa. SNF2H was detected in these fractions as two distinct isoforms, one at 135 kDa and the other
at the predicted size of 120 kDa (black arrows). Lower: IP of SNF2H from CECR2
Gt
peak fractions 3941, followed by
Western blot as in (b).
Fig. 5. Cecr2
Gt
mutants exhibit significantly reduced expression of Dlx5. qRT-PCR analysis of the indicated genes using
cDNA derived from total testis RNA. Shown are the average expression fold changes in Cecr2
Gt
mutant males normalized
to wild type (n= 3 males per genotype); error bars are standard errors of the mean. Dlx5 expression is reduced by 35%;
asterisk indicates significance (p=0.0249, two-tailed T-test).
801CECR2 Is Involved in Spermatogenesis
Author's personal copy
function in both early development and later in the
gonads that these genes may be regulated by CECR2
at both time points. We considered strong candi-
dates based primarily on their fold changes in
Cecr2
Gt
mutant E8.5 embryos and, secondly, wheth-
er they had known phenotypes affecting spermato-
genesis and/or fertility. These criteria established 12
candidates: Peg3,Dlx5,Asb4,Met,Alx1,Ndn,Amph,
Unc5c,Aspm,Rad23b,Wwtr1, and Igf1r (Fig. 5).
Quantitative RT-PCR (qRT-PCR) analysis showed
that 11 of the candidates exhibited no significant
differences in expression in Cecr2
Gt
mutants com-
pared to wild type. However, we observed a
significant 35% reduction in the expression of
Dlx5 in Cecr2 mutants (Fig. 5;p= 0.0249), suggesting
that loss of CECR2 may influence expression of Dlx5
in the germ line.
Discussion
CECR2 contributes to sperm fertility and may
regulate Dlx5 in the testis
CECR2 plays important roles in neurulation and
inner ear development,
10,19
but its functional roles in
adult tissues have not been explored. In the present
study, we found that CECR2 influences the ability of
spermatozoa to fertilize oocytes. The oocytes that are
successfully fertilized by mutant sperm correlate to
formation of blastocysts in culture and presumably
develop into live pups in vivo. Strikingly, while Cecr2
is expressed during the development of the germ cells
in the testis (Fig. 1b), the explanation for the reduced
fertility is not due to gross morphological defects in
the seminiferous epithelium resulting in aberrant
spermatogenesis. Cecr2 mutants did not show obvi-
ous defects of spermatogenesis, such as reduced
spermatid-to-spermatogonia ratios, tubule morphol-
ogy, and counts and motility of mature spermatozoa
(Fig. 2 and Table 2). Since Cecr2 is expressed strongly
in spermatogonia, with reduced levels detected in
spermatocytes and was undetectable in spermatozoa
(Fig. 1b), it is likely that it influences gene expression
and/or chromatin structure in spermatogonia, which
is then transmitted to mature spermatozoa. We
hypothesized that CECR2 may contribute to the
regulation of a panel of candidate genes in the testis,
and we observed a significant reduction of 35% in
the expression of Dlx5 in Cecr2 mutant males (Fig. 5).
Dlx5 encodes an essential homeobox transcription
factor involved in craniofacial, nervous system, and
limb development
24
and is also misregulated in
Cecr2
Gt
mutant E8.5 embryos.
18
Interestingly, Dlx5
is expressed in the fetal testis at E12.5 onward and
DLX5 partners with the transcription factor GATA-4
to regulate StAR expression and control testicular
steroidogenesis in Leydig cells.
25
We did not observe
Cecr2 expression in Leydig cells at E16.5 (Fig. 1b), but
Cecr2 is expressed in adult Leydig cells. Although
Dlx5 is known to be expressed in adult testes,
26
the
specific cell type it is expressed in is not known. The
reduced expression of Dlx5 may be an indirect result
of misregulated gene expression during testis devel-
opment in Cecr2 mutants; however, it would be
interesting to determine whether DLX5 has a non-
steroidogenic function in adult germ cells.
One possibility is that Cecr2 mutant sperm may
have a general defect in penetration of the zona
pellucida of oocytes, and therefore, CECR2 may
contribute to the expression of enzymes in the
acrosome reaction or capacitationof sperm via
cAMP signaling pathways.
27
In addition to gene
regulation, CECR2 may also contribute to chromatin
structure changes during spermatogenesis, such as
deposition of protamines or testis-specific histone
variants such as histone H1t or H3t. Further
examination of gene expression, DNA methylation,
histone modifications and variants, and protamines
in Cecr2 mutant testes would confirm whether the
biological consequences of the loss of CECR2
involve additional changes to global chromatin
structure. In addition, use of microarray or RNA-
seq to look at expression differences between wild-
type and mutant testes may reveal gene pathways
involved in the fertility defect. The use of a stronger
mutant allele, such as a conditional knockout of
Cecr2 in the germ line rather than a gene trap, would
reveal whether there are more severe fertility defects
caused by the loss of CECR2.
CECR2 forms a complex with SNF2H in the testis
Biochemical analysis of CECR2 in the testis
indicated that the mutant reporter protein CECR2
Gt
,
which lacks the bromodomain and remaining
portion of CECR2, can form complexes with ISWI
proteins, presumably due to the presence of the
DDT domain (Fig. 1a). This result is consistent with
the previous studies showing that the DDT domain
is important in the binding of Drosophila ACF1 to
ISWI
20
and of human RSF1 to SNF2H.
21
Interest-
ingly, despite observing physical interactions be-
tween CECR2
Gt
and SNF2H, we generally do not
observe reduced litter size phenotypes in Cecr2
Gt
heterozygotes (data not shown), suggesting that the
inclusion of this mutant protein into ISWI complexes
does not result in a dominant-negative effect, at least
with respect to fertility. CECR2
Gt
interacted with
SNF2H rather than SNF2L in the adult testis and
also formed a complex with SNF2H in ES cells (Fig.
4). Although these data will require confirmation
with wild-type CECR2 to rule out potential artifacts
of the mutant reporter, preferential binding to
SNF2H over SNF2L has been previously demon-
strated for other ISWI-binding proteins such as
WSTF/BAZ1B and ACF1/BAZ1A.
5,6
Consistent
802 CECR2 Is Involved in Spermatogenesis
Author's personal copy
with SNF2H being the major ISWI binding partner
of CECR2 in the testis, Cecr2 expression patterns are
similar to the expression pattern of Smarca5 (SNF2H)
which is also strongly expressed in spermatogonia
and spermatocytes, whereas in contrast, Smarca1
(SNF2L) expression is dispersed throughout the
seminiferous epithelium and mature spermatids.
2,28
Thus, in tissues undergoing extensive proliferation
such as the epiblast of the expanded blastocyst or the
seminiferous epithelium, CECR2 may interact with
SNF2H, which is associated with proliferating cell
types in the nervous system.
2
In contrast, in cell
types that have undergone differentiation such as
HEK293, CECR2 may interact predominantly with
SNF2L.
10
Together, these observations suggest that
CECR2-containing complexes exhibit cell type spec-
ificity with regard to the ISWI subunit.
CERF purified from HEK293 cells is 600 kDa,
likely consisting of a heterotetramer of two CECR2
subunits and two SNF2L subunits.
10
In contrast, the
CECR2
Gt
/SNF2H-containing complex was 0.9
1 MDa in testis (Fig. 4c), suggesting that it may
include other subunits or that it may include more
CECR2 and SNF2H subunits compared to CERF. In
addition, the CECR2
Gt
/SNF2H-containing complex
in ES cells eluted over a broad range of sizes, with a
peak at 300400 kDa, consistent with the size of a
CECR2/SNF2H heterodimer, but also up to
700 kDa approximating the size of a heterotetra-
mer (Fig. 4e). Although the difference in inferred
subunit numbers may be due to the inclusion of the
mutant CECR2 reporter protein, it may also be due
to differences in the cell types. In the testis, the
mutant CECR2
Gt
-containing complex appeared to
assemble independently of wild-type CECR2 com-
plexes, and the incorporation of SNF2H into other
ISWI complexes was unperturbed in Cecr2 mutants
(Fig. 4d and e). Whether or not all the of the CECR2
proteins in the germ line function solely in the
context of a SNF2H-containing complex is unclear
and will require further characterization of CECR2-
containing complexes in the testis by mass spec-
trometry. The role of SNF2H in the male germ line
has not been investigated; however, hypomorphic
mutation of Smarca5 was reported to affect epige-
netic repression of the variegating locus Agouti viable
yellow specifically in the male germ line,
28
suggest-
ing that SNF2H may have functional roles in male
germ cells. Thus, it is likely that, at least, some of the
functions of CECR2 in spermatogenesis would be in
the context of a complex with SNF2H.
Conclusion
Here, we report that the ISWI binding partner
CECR2 contributes to male fertility in a manner that
usually does not drastically alter seminiferous
epithelium structure, sperm count, motility, or
morphology but nevertheless causes defects in
oocyte fertilization. This is to our knowledge the
first report on a chromatin remodeling protein with
this particular mutant phenotype. One model in
support of our findings is that the mutant CECR2
Gt
protein can form a complex with SNF2H and
possibly other proteins as would be expected of
wild-type CECR2, and the complex can localize to
the nucleus but is not functional. In Cecr2
Gt
heterozygotes, both types of complexes may be
present, but the mutant CECR2
Gt
complexes do not
interfere with the wild-type ones, resulting in
sufficient levels of functional CECR2. In Cecr2
Gt
mutants, the resulting deficiency in chromatin
remodeling activity and/or targeting may result in
misregulated gene expression and/or deleterious
changes in chromatin structure, which cause fertility
defects in the mature spermatozoa.
Materials and Methods
Cecr2
Gt45Bic
mutation, genotyping, and fertility
analysis
All mice used in this study were housed at the Biological
Sciences Animal Services facility at the University of
Alberta in Edmonton, Canada. The Cecr2
Gt45Bic
gene trap
mutation on BALB/c background (Cecr2
Gt
) was generated
previously and genotyped by multiplex PCR.
10,19
For our
initial fertility testing (see Fig. 1), 11 BALB/c Cecr2
Gt45Bic
mutant males and 23 wild-type males between ages of 10
and 24 weeks were each set up in mating pairs to wild-type
females and left together for 3 months. If no pups were
sired within the normal 3- to 4-week period, a new female
was provided. The number of pups was counted in each
litter, and litter sizes for all wild-type and Cecr2 mutant
males were averaged. For the second fertility analysis
(Tables 1,2, and 3), BALB/c wild-type and Cecr2
Gt45Bic
homozygous mutants males (non-penetrant for exence-
phaly) aged 6 weeks and older were housed with a one or
two wild-type BALB/c females. Females were subsequent-
ly tested for seminal plugs, and pregnancies were timed
such that all plugged females were euthanized and
dissected between E14.5 and E18.5. The numbers of
morphologically normal and undeveloped embryos
(resorbed) were counted in each litter. Males were given
2 days apart from females and then again housed with
wild-type females for another round of mating. All
procedures involving mice were carried out in accordance
with the Animal Care and Use Committee at the University
of Alberta and national guidelines and policies.
Culture of CT45 mouse embryonic stem cells
The CT45 ES cell line was generated in a previous
study
23
and was kindly provided by Dr. W. Bickmore. ES
cells were maintained in an undifferentiated state in
DMEM (Dulbecco's modified Eagle's medium) high-
glucose media containing 15% fetal bovine serum, 2 mM
L-glutamine, 0.1 mM nonessential amino acids, 100 mM β-
803CECR2 Is Involved in Spermatogenesis
Author's personal copy
mercaptoethanol, 100 U/ml penicillin, and 100 mg/ml
streptomycin (all from Invitrogen), with 1000 U/ml
leukemia inhibitory factor (Sigma-Aldrich), and were
cultured on gelatinized plates in a 5% CO
2
atmosphere
at 37 °C.
Histology, Xgal staining of testes, and spermatozoa
analysis
Xgal staining of the Cecr2
Gt45Bic
gene product was
performed as previously described.
10,19
For histology,
testes were dissected, fixed in 4% paraformaldehyde,
sectioned at 5 μm, and mounted on positively charged
slides. Sections were stained with hematoxylin and eosin
and imaged according to previous methods.
19
Spermatid-
to-spermatogonia ratios were determined for four BALB/c
wild-type males and five Cecr2
Gt45Bic
mutant males by
counting spermatids and spermatogonia in 10 sections of
the seminiferous epithelium of each animal (blind to
genotype), and average ratios were recorded. Analyses of
sperm count, motility, and morphology were performed as
previously described.
29
Mature sperms isolated from the
cauda epididymis of wild-type and Cecr2
Gt45Bic
mutant
males were counted by hemocytometer in RPMI media
(Sigma-Aldrich). For analysis of motility and morphology,
sperm was isolated as above and examined on a slide
under a Nikon TMS inverted microscope. Approximately
200 sperms were scored for each male under four
specific categories describing its motility pattern: (1)
moving forward, (2) moving in any direction except
forward, (3) moving but not advancing in any direction,
and (4) nonmotile. Approximately 200 sperms from each
male were scored for their morphology as linear/
normal, angulated (bent at N90° or hairpin shaped), or
headless.
Superovulation and analysis of fertilization and
preimplantation embryos
FVB/N wild-type females were superovulated using
previous methods with minor modifications.
30
Females
were injected with 5 IU pregnant mare serum gonadotro-
pin/Folligon followed by 5 IU human chorionic gonado-
tropin/Chorulon (Sigma-Aldrich/Intervet) approximately
46 h later. One superovulated female was then housed
with a BALB/c wild-type or Cecr2
Gt45Bic
mutant male for
approximately 16 h, and females were plug-tested.
Plugged females were euthanized and dissected, and
ovaries were disrupted to release the oocytes into M2
media (Sigma-Aldrich) containing 0.3 mg/ml hyaluroni-
dase (Sigma-Aldrich) for 1.5 min to digest the cumulus.
Oocytes were then washed in M2 media and viewed by
phase-contrast microscopy using the Axioscope II plus
(Carl Zeiss Canada Ltd.). The number of fertilized oocytes
from each superovulated female was scored by visualizing
the maternal and paternal pronuclei. Oocytes were then
washed twice in M16 media (Sigma-Aldrich) and cultured
in 20 μl of M16 media containing 60 mg/l penicillin and
50 mg/l streptomycin (Invitrogen) in microdrop plates
covered with mineral oil at 37 °C and 5% CO
2
atmosphere.
Development of zygotes was observed each day, and the
number of surviving blastocysts at approximately E3.5-4
was scored.
RT-PCR and qRT-PCR analyses
For RT-PCR, total RNA was extracted from ES cells
and adult testes using the Qiagen RNeasy lipid tissue
mini kit according to the product protocol, and 1 μgof
RNA was used for cDNA synthesis using the Super-
Script III first-strand cDNA synthesis kit from Invitro-
gen according to the manufacturer's instructions. cDNA
synthesis was performed using 2 pmol of βgeo reverse
primer 5AAATTCAGACGGCAAACGAC 3. Negative
controls were performed by omitting RT from the cDNA
synthesis reaction. The following primers were used for
PCR amplification of Cecr2 cDNA spanning from exon 1
to exon 7: forward 5GAGCGAGAGCGAGTGAGC 3
and reverse 5CTCGGAAGCTCTCAGTGACC 3. PCR
was performed with an initial denaturing step of 95 °C
for 2 min, followed by 30 cycles of the following: 95 °C
for 15 s, 55 °C for 30 s, and 68 °C for 1 min; a final
extension was performed at 68 °C for 3 min. Sequencing
of cDNA was performed by Sanger dideoxy sequencing.
qRT-PCR analysis was performed as previously
described.
18
Briefly, 1 μg of total RNA each from testes
isolated from three wild-type and Cecr2
Gt
mutant males
was converted into cDNA using the SuperScript VILO
kit (Invitrogen), and quantitative PCR was performed
using Taq Universal PCR Master Mix (Applied
Biosystems) on the Applied Biosystems StepOne Plus
thermocycler with three technical replicates per sample
for each target. Fold changes were calculated by delta
delta C
T
method using Elmo2 as an endogenous
control. Primer-probe sequences are available upon
request.
Western blot, immunoaffinity enrichment, and
gel filtration
Nuclear extracts were prepared from ES cells and
mouse tissues according to previous methods.
31,32
Protein
concentration was determined by the DC (detergent
compatible) protein assay (Bio-Rad). For Western blot
analysis, Tris glycine SDS-PAGE was performed followed
by transfer to polyvinylidene fluoride membranes. Mem-
branes were blocked with 5% skim milk in Tris-buffered
saline [25 mM Tris (pH 8.0) and 150 mM NaCl] containing
0.05% Tween-20 and were incubated with primary
antibodies overnight at 4 °C. Antibodies used for Western
blot were as follows: anti-β-galactosidase (Santa Cruz
Biotechnology), anti-SNF2H (Active Motif), anti-TATA-
binding protein (Sigma-Aldrich), anti-SNF2L (gift from
Dr. D. Picketts), anti-ACF1 (gift from Dr. P. Varga-Weisz),
anti-WSTF (gift from Dr. P. Varga-Weisz), anti-RSF1 (gift
from Dr. D. Reinberg), and anti-TIP5 (gift from Dr. I.
Grümmt). Primary antibodies were detected using horse-
radish-peroxidase-conjugated anti-rabbit, anti-mouse, and
anti-goat IgG secondary antibodies (Sigma-Aldrich).
Bands were visualized by ECL plus (GE Healthcare) and
exposure to X-ray film.
For immunoaffinity enrichment of CECR2
Gt
,0.5 mg
monoclonal β-galactosidase antibodies (Meridian Life
Science) was cross-linked to 0.5 ml protein G-Sepharose
using previous methods.
33
Nuclear extract from Cecr2
Gt45Bic
heterozygous adult testes or CT45 ES cells (4 mg) was
diluted to a final concentration of 210 mM NaCl and first
804 CECR2 Is Involved in Spermatogenesis
Author's personal copy
passed over a mock column containing no antibody at
gravity flow, then over the β-galactosidase antibody
column. Bound proteins on each column were washed
extensively with wash buffer [20 mM Hepes (pH 7.9),
400 mM NaCl, 0.1% Triton-X-100, and 10% glycerol] and
stepwise eluted by the addition of 0.2 M glycine (pH 2.5).
Fractions were neutralized by the addition of 1.5 M Tris
(pH 8.8) and were analyzed by Western blot.
Gel-filtration analysis was performed as previously
described.
34
A column of Sephacryl S-400 HR (GE
Healthcare) was equilibrated in 20 mM Hepes (pH 7.9),
420 mM NaCl, 1.5 mM MgCl
2
, 0.2 mM ethylenediamine-
tetraacetic acid, and 10% glycerol and was calibrated
using purified standards blue dextran (2 MDa), thyro-
globulin (670 kDa), and bovine serum albumin (67 kDa)
(all from Sigma-Aldrich). Nuclear extracts (23 mg)
were fractionated over the column at a flow rate of 0.5
0.8 ml/min, and 0.5-ml fractions were collected and
either stored at 20 °C or precipitated with trichloroacetic
acid, washed with acetone, and air-dried. Protein pellets
were dissolved in SDS-PAGE loading buffer and ana-
lyzed by Western blot. For analysis of DNA-dependent
interactions during gel filtration, ethidium bromide was
added to the nuclear extract at 50 μg/ml and incubated
for 1 h on ice before fractionation as previously
described.
35
IP was performed from gel-filtration frac-
tions essentially as described previously.
36
The three
peak CECR2
Gt
-containing fractions as judged by Western
blot were pooled, diluted 1:2, and separated into two
volumes, and IP was performed overnight at 4 °C on each
volume in parallel by adding 2 μl of anti-SNF2H (Active
Motif) or 2 μg of mouse IgG (Sigma-Aldrich) in IP buffer
[20 mM Hepes (pH 7.9), 210 mM NaCl, 0.2 mM
ethylenediaminetetraacetic acid, 0.1% Triton-X-100, and
10% glycerol] containing 1 mM phenylmethyl sulfonyl
fluoride. Immunocomplexes were collected by adding
30 μl protein A/G-agarose (Santa Cruz Biotechnology)
and rotating an additional 2 h at 4 °C. Beads were
washed four times with IP buffer, and bound proteins
were eluted by boiling in SDS-PAGE loading buffer.
Acknowledgements
We wish to thank Dr. D. Picketts for SNF2L
antibodies and critical reading of this manuscript.
We would also like to thank N. Fairbridge and Dr.
Michael Dyck for helpful discussions. We thank Dr.
W. A. Bickmore for the CT45 ES cell line, Dr. P.
Varga-Weisz for ACF1 and WSTF antibodies, Dr. D.
Reinberg for RSF1 antibodies, and Dr. I. Grümmt for
TIP5 antibodies. This work was supported by the
Canadian Institutes of Health Research Grant
MOP64361. P.J.T. was supported by a Province of
Alberta Queen Elizabeth II scholarship. K.A.N. was
supported by a summer studentship from Alberta
Innovates Health Solutions. C.E.D. was supported
by a scholarship from the Natural Sciences and
Engineering Research Council of Canada. We
declare no conflicts of interest.
References
1. Ho, L. & Crabtree, G. R. (2010). Chromatin remodel-
ling during development. Nature,463, 474484.
2. Lazzaro, M. A. & Picketts, D. J. (2001). Cloning and
characterization of the murine Imitation Switch (ISWI)
genes: differential expression patterns suggest distinct
developmental roles for Snf2h and Snf2l. J. Neurochem.
77, 11451156.
3. LeRoy, G., Orphanides, G., Lane, W. S. & Reinberg, D.
(1998). Requirement of RSF and FACT for transcrip-
tion of chromatin templates in vitro.Science,282,
19001904.
4. LeRoy, G., Loyola, A., Lane, W. S. & Reinberg, D.
(2000). Purification and characterization of a human
factor that assembles and remodels chromatin. J. Biol.
Chem. 275, 1478714790.
5. Poot, R. A., Dellaire, G., Hulsmann, B. B., Grimaldi,
M. A., Corona, D. F., Becker, P. B. et al. (2000).
HuCHRAC, a human ISWI chromatin remodelling
complex contains hACF1 and two novel histone-fold
proteins. EMBO J. 19, 33773387.
6. Bozhenok, L., Wade, P. A. & Varga-Weisz, P. (2002).
WSTFISWI chromatin remodeling complex targets
heterochromatic replication foci. EMBO J. 21,
22312241.
7. Strohner, R., Nemeth, A., Jansa, P., Hofmann-Rohrer,
U., Santoro, R., Langst, G. & Grummt, I. (2001).
NoRCa novel member of mammalian ISWI-con-
taining chromatin remodeling machines. EMBO J. 20,
48924900.
8. Bochar, D. A., Savard, J., Wang, W., Lafleur, D. W.,
Moore, P., Cote, J. & Shiekhattar, R. (2000). A family of
chromatin remodeling factors related to Williams
syndrome transcription factor. Proc. Natl Acad. Sci.
USA,97, 10381043.
9. Barak, O., Lazzaro, M. A., Lane, W. S., Speicher, D. W.,
Picketts, D. J. & Shiekhattar, R. (2003). Isolation of
human NURF: a regulator of Engrailed gene expres-
sion. EMBO J. 22, 60896100.
10. Banting, G. S., Barak, O., Ames, T. M., Burnham, A. C.,
Kardel, M. D., Cooch, N. S. et al. (2005). CECR2, a
protein involved in neurulation, forms a novel
chromatin remodeling complex with SNF2L. Hum.
Mol. Genet. 14, 513524.
11. Collins, N., Poot, R. A., Kukimoto, I., Garcia-Jimenez,
C., Dellaire, G. & Varga-Weisz, P. D. (2002). An
ACF1ISWI chromatin-remodeling complex is re-
quired for DNA replication through heterochromatin.
Nat. Genet. 32, 627632.
12. Poot, R. A., Bozhenok, L., van den Berg, D. L.,
Steffensen, S., Ferreira, F., Grimaldi, M. et al. (2004).
The Williams syndrome transcription factor interacts
with PCNA to target chromatin remodelling by ISWI
to replication foci. Nat. Cell Biol. 6, 12361244.
13. Zhou, Y., Santoro, R. & Grummt, I. (2002). The
chromatin remodeling complex NoRC targets
HDAC1 to the ribosomal gene promoter and represses
RNA polymerase I transcription. EMBO J. 21,
46324640.
14. Landry, J., Sharov, A. A., Piao, Y., Sharova, L. V., Xiao,
H., Southon, E. et al. (2008). Essential role of chromatin
remodeling protein Bptf in early mouse embryos and
embryonic stem cells. PLoS Genet. 4, e1000241.
805CECR2 Is Involved in Spermatogenesis
Author's personal copy
15. Perpelescu, M., Nozaki, N., Obuse, C., Yang, H. & Yoda,
K. (2009). Active establishment of centromeric CENP-A
chromatin by RSF complex. J. Cell Biol. 185,397407.
16. Stopka, T. & Skoultchi, A. I. (2003). The ISWI ATPase
Snf2h is required for early mouse development. Proc.
Natl Acad. Sci. USA,100, 1409714102.
17. Lazzaro, M. A., Pepin, D., Pescador, N., Murphy,
B. D., Vanderhyden, B. C. & Picketts, D. J. (2006). The
imitation switch protein SNF2L regulates steroido-
genic acute regulatory protein expression during
terminal differentiation of ovarian granulosa cells.
Mol. Endocrinol. 20, 24062417.
18. Fairbridge, N. A., Dawe, C. E., Niri, F. H., Kooistra,
M. K., King-Jones, K. & McDermid, H. E. (2010). Cecr2
mutations causing exencephaly trigger misregulation
of mesenchymal/ectodermal transcription factors.
Birth Defects Res., Clin. Mol. Teratol. 88, 619625.
19. Dawe, C. E., Kooistra, M. K., Fairbridge, N. A., Pisio,
A. C. & McDermid, H. E. (2011). Role of chromatin
remodeling gene Cecr2 in neurulation and inner ear
development. Dev. Dyn. 240, 372383.
20. Fyodorov, D. V. & Kadonaga, J. T. (2002). Binding of
Acf1 to DNA involves a WAC motif and is important
for ACF-mediated chromatin assembly. Mol. Cell. Biol.
22, 63446353.
21. Sheu, J. J., Choi, J. H., Yildiz, I., Tsai, F. J., Shaul, Y.,
Wang, T. L. & Shih Ie, M. (2008). The roles of human
sucrose nonfermenting protein 2 homologue in the
tumor-promoting functions of Rsf-1. Cancer Res. 68,
40504057.
22. Aravind, L. & Landsman, D. (1998). AT-hook motifs
identified in a wide variety of DNA-binding proteins.
Nucleic Acids Res. 26, 44134421.
23. Tate, P., Lee, M., Tweedie, S., Skarnes, W. C. &
Bickmore, W. A. (1998). Capturing novel mouse genes
encoding chromosomal and other nuclear proteins.
J. Cell Sci. 111, 25752585.
24. Depew, M. J., Liu, J. K., Long, J. E., Presley, R., Meneses,
J. J., Pedersen, R. A. & Rubenstein, J. L. (1999). Dlx5
regulates regional development of the branchial arches
and sensory capsules. Development,126, 38313846.
25. Nishida, H., Miyagawa, S., Vieux-Rochas, M., Morini,
M.,Ogino,Y.,Suzuki,K.et al. (2008). Positive
regulation of steroidogenic acute regulatory protein
gene expression through the interaction between Dlx
and GATA-4 for testicular steroidogenesis. Endocri-
nology,149, 20902097.
26. Kimura, M. I., Kazuki, Y., Kashiwagi, A., Kai, Y., Abe,
S., Barbieri, O. et al. (2004). Dlx5, the mouse
homologue of the human-imprinted DLX5 gene, is
biallelically expressed in the mouse brain. J. Hum.
Genet. 49, 273277.
27. Fraser, L. R. (2010). The switching onof mammalian
spermatozoa: molecular events involved in promotion
and regulation of capacitation. Mol. Reprod. Dev. 77,
197208.
28. Chong, S., Vickaryous, N., Ashe, A., Zamudio, N.,
Youngson, N., Hemley, S. et al. (2007). Modifiers of
epigenetic reprogramming show paternal effects in
the mouse. Nat. Genet. 39, 614622.
29. Netzel-Arnett, S., Bugge, T. H., Hess, R. A., Carnes, K.,
Stringer, B. W., Scarman, A. L. et al. (2009). The
glycosylphosphatidylinositol-anchored serine prote-
ase PRSS21 (testisin) imparts murine epididymal
sperm cell maturation and fertilizing ability. Biol.
Reprod. 81, 921932.
30. Eggan, K. & Jaenisch, R. (2003). Differentiation of F1
embryonic stem cells into viable male and female mice
by tetraploid embryo complementation. Methods
Enzymol. 365,2539.
31. Dignam, J. D., Lebovitz, R. M. & Roeder, R. G. (1983).
Accurate transcription initiation by RNA polymerase
II in a soluble extract from isolated mammalian nuclei.
Nucleic Acids Res. 11, 14751489.
32. Cox, B. & Emili, A. (2006). Tissue subcellular
fractionation and protein extraction for use in mass-
spectrometry-based proteomics. Nat. Protoc. 1,
18721878.
33. Chalkley, G. E. & Verrijzer, C. P. (2004). Immuno-
depletion and purification strategies to study chro-
matin-remodeling factors in vitro.Methods Enzymol.
377, 421442.
34. Lewis, P. W., Beall, E. L., Fleischer, T. C., Georlette, D.,
Link, A. J. & Botchan, M. R. (2004). Identification of a
Drosophila Myb-E2F2/RBF transcriptional repressor
complex. Genes Dev. 18, 29292940.
35. Hanai, K., Furuhashi, H., Yamamoto, T., Akasaka, K.
& Hirose, S. (2008). RSF governs silent chromatin
formation via histone H2Av replacement. PLoS Genet.
4, e1000011.
36. Cavellan, E., Asp, P., Percipalle, P. & Farrants, A. K.
(2006). The WSTFSNF2h chromatin remodeling
complex interacts with several nuclear proteins in
transcription. J. Biol. Chem. 281, 1626416271.
806 CECR2 Is Involved in Spermatogenesis
... Epigenetic aberrations play critical roles in driving breast cancer metastasis and may be reversibly regulated by targeting epigenetic regulators (4,(19)(20)(21)(22)(23). Cat eye syndrome chromosome region candidate 2 (CECR2) is an epigenetic factor with a bromodomain that recognizes acetylated lysine residues. CECR2 was shown to play critical roles in DNA damage responses (24), neurulation (25), and spermatogenesis (26). It was reported that CECR2 participates in chromatin remodeling by interacting with sucrose nonfermenting 2 like and homolog (SNF2L and SNF2H, respectively) (25,26). ...
... CECR2 was shown to play critical roles in DNA damage responses (24), neurulation (25), and spermatogenesis (26). It was reported that CECR2 participates in chromatin remodeling by interacting with sucrose nonfermenting 2 like and homolog (SNF2L and SNF2H, respectively) (25,26). The bromodomain of CECR2 was predicted to be highly druggable (27). ...
... CECR2 was reported to form chromatin remodeling complexes with SNF2L or SNF2H; thus, it may affect chromatin accessibility (25,26). We therefore performed assay for transposase-accessible chromatin using sequencing (ATAC-seq) of CECR2 knockout and control LM2 cells. ...
Article
Full-text available
Metastasis is the major cause of cancer-related deaths due to the lack of effective therapies. Emerging evidence suggests that certain epigenetic and transcriptional regulators drive cancer metastasis and could be targeted for metastasis treatment. To identify epigenetic regulators of breast cancer metastasis, we profiled the transcriptomes of matched pairs of primary breast tumors and metastases from human patients. We found that distant metastases are more immune inert with increased M2 macrophages compared to their matched primary tumors. The acetyl-lysine reader, cat eye syndrome chromosome region candidate 2 (CECR2), was the top up-regulated epigenetic regulator in metastases associated with an increased abundance of M2 macrophages and worse metastasis-free survival. CECR2 was required for breast cancer metastasis in multiple mouse models, with more profound effect in the immunocompetent setting. Mechanistically, the nuclear factor κB (NF-κB) family member v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA) recruits CECR2 to increase chromatin accessibility and activate the expression of their target genes. These target genes include multiple metastasis-promoting genes, such as TNC, MMP2, and VEGFA, and cytokine genes CSF1 and CXCL1, which are critical for immunosuppression at metastatic sites. Consistent with these results, pharmacological inhibition of CECR2 bromodomain impeded NF-κB-mediated immune suppression by macrophages and inhibited breast cancer metastasis. These results reveal that targeting CECR2 may be a strategy to treat metastatic breast cancer.
... Unlike other BRDs, BRD and extraterminal (BET) proteins have two tightly packed BRDs that specifically bind diacetylated lysine residues (25,26), promote chromatin opening, recruit transcription factors and coactivators to target gene promoters and enhancers, and activate the RNA polymerase II (Pol II) complex to promote transcriptional elongation (27). Similarly, tandem plant homeodomain (PHD) fingers with BRDs contribute to the assembly and activity of their associated complexes in nucleosomes and act on DNA replication in chromosome segregation (28,29). BRDs are widely involved in the transcription of cancer-related genes, and depending on the subtle differences in BRDs and various combinations of functional groups, highly selective inhibitors may be designed with potential clinical applications for the treatment of metastatic cancers (30). ...
Article
Full-text available
Histone alterations are a hallmark of kidney cancer. Histone acetylation modification mediated by bromodomain proteins (BRD) has been indicated to be related to a variety of cancer types and several targeted inhibitors have been proven to be promising modalities for cancer adjuvant therapy. As renal cell carcinoma (RCC) is not sensitive to radiotherapy or chemotherapy, the exploration of effective adjuvant therapies remains an important research direction for advanced RCC. At present, studies on bromodomain family proteins in RCC are limited and the roles of bromodomain family proteins in RCC have remained to be fully elucidated. The present review discussed the role of bromodomain family proteins in RCC, aiming to explore possible potential therapeutic targets of BRD‑related drugs in this type of cancer.
... They also play roles in DNA replication of the heterochromatin component [27••]. The CECR2 protein is known to contribute to spermatogenesis [63] and DNA damage response [64]. ...
Article
Full-text available
Purpose of Review An understanding of epigenetic marks on chromatin and their roles in development and disease has been a highlight of the past few years of biological research. Acetylated lysine is one such chromatin mark that is specifically read by modules called bromodomains. Proteins that harbor bromodomain modules have been implicated in cancer and other disease conditions and developmental abnormalities. Recent Findings This review looks into recent findings about all known bromodomain-containing proteins in higher eukaryotes based on their family and enzymatic function. It further evaluates the consequences of mutations in these proteins resulting in diseased conditions. Finally, this review delves into potential bromodomain-based therapeutics that are under investigation or approved by the FDA. Summary Epigenetic readers such as bromodomains that are present in few mammalian proteins impart them the ability to identify acetylated lysine marks on chromatin and functionally translate them into gene expression or repression affecting overall cellular and tissue phenotypes. We have summarized the latest updates in this field.
... were expressed higher in testes than in ovaries, suggesting they were potentially involved in testis development and the maintenance of spermatogenesis. For example, cecr2 was highly expressed in adult spermatogonia, and disruption of cecr2 led to compromised ability to fertilize oocytes in the male mouse [66]. Ep300a was also reported to be expressed higher in the testis tissue of the medaka and Mexican tetra [67]. ...
Article
Full-text available
The bromodomain (BRD) proteins specifically recognize the N-acetyllysine motifs, which is a key event in the reading process of epigenetic marks. BRDs are evolutionarily highly conserved. Over recent years, BRDs attracted great interest because of their important roles in biological processes. However, the genome-wide identification of this family was not carried out in many animal groups, in particular, in teleosts. Moreover, the expression patterns were not reported for any of the members in this family, and the role of the BRD family was not extensively studied in fish reproduction. In this study, we identified 16 to 120 BRD genes in 24 representative species. BRDs expanded significantly in vertebrates. Phylogenetic analysis showed that the BRD family was divided into eight subfamilies (I–VIII). Transcriptome analysis showed that BRDs in Nile tilapia (Oreochromis niloticus) exhibited different expression patterns in different tissues, suggesting that these genes may play different roles in growth and development. Gonadal transcriptome analysis showed that most of the BRDs display sexually dimorphic expression in the gonads at 90 and 180 dah (days after hatching), including 21 testis-dominated genes (brdt, brd4a and brd2b, etc.), and nine ovary-dominated genes (brd3b, brd2a and kat2a, etc.). Consistent with transcriptomic data, the results of qRT-PCR and fluorescence in situ hybridization showed that brdt expression was higher in the testis than in the ovary, suggesting its critical role in the spermatogenesis of the tilapia. Male fish treated with JQ1 (BET subfamily inhibitor) displayed abnormal spermatogenesis. The numbers of germ cells were reduced, and the expression of steroidogenic enzyme genes was downregulated, while the expression of apoptosis-promoting genes was elevated in the testis tissue of treated fish. Our data provide insights into the evolution and expression of BRD genes, which is helpful for understanding their critical roles in sex differentiation and gonadal development in teleosts.
... Except for CERF (4) and NURF (6), which contain SMARCA1, SMARCA5 is present in all ISWI complexes. This ATPase variability may be largely cell type specific (68). ...
Article
Full-text available
During aging, cells are continuously exposed to a range of environmental and endogenous conditions causing DNA damage, genomic instability and cellular stresses. At the DNA damage sites, essential biological processes such as transcription and replication are impeded, which can cause diseases of premature aging and cancer. Chromatin structure limits access to DNA, which serves as a docking site for DNA repair, signaling proteins and transcription factors. There is growing body of evidence that suggests that chromatin structure is involved in regulating aging. Accordingly, the activity of ATP-dependent chromatin remodeling complexes is gradually gaining interest in this process. Despite ATP-dependent chromatin remodelers being extensively studied, their role in aging and lifespan regulation remains less studied. This review focuses mainly on discussing the role of ATP-dependent chromatin remodelers in genome stability, induction of stress response pathways and regulation of aging.
Article
Full-text available
Introduction: Neural tube defects (NTDs) are serious congenital malformations. The etiology of NTDs involves both genetic and environmental factors. Loss of CECR2 in mice has been shown to result in NTDs. Our previous study indicated that high homocysteine (HHcy) levels could further reduced the expression level of CECR2. This investigation aims to explore the genetic influence of the chromatin remodeling gene, CECR2, in humans and determine if HHcy can have a synergistic effect on protein expression. Methods: We conducted Next-Generation Sequencing (NGS) of the CECR2 gene in 373 NTD cases and 222 healthy controls, followed by functional assay application to select and evaluate CECR2 missense variants and subsequent Western blotting to identify protein expression levels. Results: From the analysis, we identified nine rare, NTD-specific mutations within the CECR2 gene. Significantly, four missense variants (p.E327V, p.T521S, p.G701R, and p.G868R) were selected via functional screening. The E9.5 mouse ectodermal stem cell line NE-4C, transfected with plasmids expressing p.E327V, p.T521S, p.G868R variants or a recombinant harboring all four (named as 4Mut), exhibited notable reductions in CECR2 protein expression. Furthermore, exposure to homocysteine thiolactone (HTL), an extremely reactive homocysteine metabolite, amplified the reduction in CECR2 expression, accompanied by a significant increase in the apoptotic molecule Caspase3 activity, a potential NTD inducer. Importantly, folic acid (FA) supplementation effectively counteracted the CECR2 expression decline induced by CECR2 mutation and HTL treatment, leading to reduced apoptosis. Discussion: Our observations underscore a synergistic relationship between HHcy and genetic variations in CECR2 concerning NTDs, thereby reinforcing the concept of gene-environment interaction phenomena in NTD etiology.
Article
Comprehensive Summary Cat's eye syndrome chromosome candidate 2 bromodomain (CECR2 BRD) and Bromodomain PHD transcription factor bromodomain (BPTF BRD) are the same subfamily proteins, both of which are highly conserved in sequence and binding pockets. Challenges remain in the development of small molecules to inhibit one of the two bromodomains (BRDs), in view of each subtype may possess unique physiological and pathological functions. There is still a lack of effective selective inhibitors of CECR2 BRD, which makes it difficult to fully understand the pathogenesis of CECR2‐BRD in diseases, especially cancers. Herein, we report our efforts to discover a series of highly selective CECR2 BRD inhibitors over BPTF BRD based on TP‐248. Structure‐based molecular optimization led to the discovery of DC‐CEi‐26, whose IC 50 for CECR2 BRD was 96.7 ± 14.9 nmol/L and selectivity was up to 590 × over BPTF BRD. DC‐CEi‐26 showed weak potencies for other classic BRDs in different subfamily, which may serve as a chemical probe for CECR2 BRD biological research.
Article
Cat eye syndrome chromosome region candidate 2 (CECR2) bromodomain is a module of CECR2-containing remodeling factor (CERF), which is a chromatin remodeling complex correlating with transcriptional control and adjustment of chromatin architecture. Potent chemical probes would be beneficial to gain insights into the biochemical and pharmacological functions of CECR2 BRD. Herein, we report the discovery of a series of CECR2 BRD inhibitors with 7H-pyrrolo[2,3-d] pyrimidine scaffold based on molecular docking model of TP-248 and CECR2 BRD. The most potent inhibitor of this series, DC-CBi-22 with IC50 of 8.0 ± 1.4 nM against CECR2 BRD and selectivity over BPTF BRD up to 24.9-fold. The SARs were detailed according to molecular docking. DC-CBi-22 would serve as a useful chemical probe for the study of CECR2.
Article
Defects in spermatogenesis are an important cause of male infertility. Multiple aspects of spermatogenesis are controlled by chromatin remodelers, including regulating transcription. We previously described mutations in chromatin remodeling gene Cecr2 that resulted in the lethal neural tube defect exencephaly in most mutant mice, and subfertility in mice that were non-penetrant for exencephaly. Here, we show that the severity of male subfertility is dependent on age. Cecr2 GT/Del males contain two mutant alleles, one of which is hypomorphic and therefore produces a small amount of protein. These males sire the fewest pups just after sexual maturity (88% fewer than Cecr2 +/+ at P42-60) but improve with age (49% fewer than Cecr2 +/+ at P81-100), although never completely recovering to Cecr2 +/+ (wild type) levels. When young, they also have defects in testis histology, in vivo fertilization frequency, sperm number and motility, and testis weight that show similar improvement with age. Immunostaining of staged seminiferous tubules showed CECR2 in type A, In and B spermatogonia, and less in preleptotene and leptotene spermatocytes. Histological defects were first apparent in Cecr2 GT/Del testes at P24, and RNA-seq analysis revealed 387 differentially expressed genes. This included 66 genes on the X chromosome (almost double the number on any other chromosome), all more highly expressed in Cecr2 GT/Del testes. This inappropriate expression of X chromosome genes could be caused by a failure of effective meiotic sex chromosome inactivation. We identify several abnormally expressed genes that may contribute to defects in spermatogenesis at P24. Our results support a role for Cecr2 in juvenile spermatogenesis.
Article
Chromatin remodeling complexes alter chromatin structure to control access to DNA and therefore control cellular processes such as transcription, DNA replication, and DNA repair. CECR2 is a chromatin remodeling factor that plays an important role in neural tube closure and reproduction. Loss-of-function mutations in Cecr2 result primarily in the perinatal lethal neural tube defect exencephaly, with non-penetrant mice that survive to adulthood exhibiting subfertility. CECR2 forms a complex with ISWI proteins SMARCA5 and/or SMARCA1, but further information on the structure and function of the complex is not known. We therefore have identified candidate components of the CECR2-containing remodeling factor (CERF) complex in embryonic stem (ES) cells through mass spectroscopy. Both SMARCA5 and SMARCA1 were confirmed to be present in CERF complexes in ES cells and testis. However, novel proteins CCAR2 and LUZP1 are CERF components in ES cells but not testis. This tissue specificity in mice suggests these complexes may also have functional differences. Furthermore, LUZP1, loss of which is also associated with exencephaly, appears to play a role in stabilizing the CERF complex in ES cells. Keywords: CECR2, LUZP1, CCAR2, Chromatin remodeling factor, Neural tube defects.
Article
Full-text available
Mammalian chromatin remodeling complexes are involved in both activation and repression of transcription. Here, we show that NoRC, a SNF2h-containing nucleolar chromatin remodeling complex, represses ribosomal gene transcription. NoRC-mediated rDNA silencing was alleviated by trichostatin A, indicating that histone deacetylation is causally involved in silencing. Chromatin immunoprecipitation experiments demonstrate that overexpression of TIP5, the large subunit of NoRC, mediates deacetylation of nucleosomes in the vicinity of the rDNA promoter. Protein±protein interaction assays reveal association of TIP5 with the histone deacetylase HDAC1 in vivo and in vitro. Deletion of the C-terminal PHD ®nger and bromodomain abolishes the interaction of TIP5 and HDAC1, and abrogates transcriptional repression. The results suggest that NoRC silences the rDNA locus by targeting the SIN3 corepressor complex to the rDNA promoter, thereby establishing a repressed chromatin structure.
Article
Full-text available
Chromatin remodelling complexes containing the nucleosome-dependent ATPase ISWI were first isolated from Drosophila embryos (NURF, CHRAC and ACF). ISWI was the only common component reported in these complexes. Our purification of human CHRAC (HuCHRAC) shows that ISWI chromatin remodelling complexes can have a conserved subunit composition in completely different cell types, suggesting a conserved function of ISWI. We show that the human homologues of two novel putative histone-fold proteins in Drosophila CHRAC are present in HuCHRAC. The two human histone-fold proteins form a stable complex that binds naked DNA but not nucleosomes. HuCHRAC also contains human ACF1 (hACF1), the homologue of Acf1, a subunit of Drosophila ACF. The N-terminus of mouse ACF1 was reported as a heterochromatin-targeting domain. hACF1 is a member of a family of proteins with a related domain structure that all may target heterochromatin. We discuss a possible function for HuCHRAC in heterochromatin dynamics. HuCHRAC does not contain topoisomerase II, which was reported earlier as a subunit of Drosophila CHRAC.
Article
Full-text available
Rsf-1 interacts with human sucrose nonfermenting protein 2 homologue (hSNF2H) to form a chromatin remodeling complex that participates in several biological processes. We have previously shown that Rsf-1 gene amplification was associated with the most aggressive type of ovarian cancer and cancer cells with Rsf-1 overexpression depended on Rsf-1 to survive. In this report, we determine if formation of the Rsf-1/hSNF2H complex could be one of the mechanisms contributing to tumor cell survival and growth in ovarian carcinomas. Based on immunohistochemistry, we found that Rsf-1 and hSNF2H were co-upregulated in ovarian cancer tissues. Ectopic expression of Rsf-1 in SKOV3 ovarian cancer cells with undetectable endogenous Rsf-1 expression enhanced hSNF2H protein levels and promoted SKOV3 tumor growth in a mouse xenograft model. Our studies also indicated that induction of Rsf-1 expression affected the molecular partnership of hSNF2H and translocated hSNF2H into nuclei where it colocalized with Rsf-1. Furthermore, analysis of Rsf-1 deletion mutants showed that the Rsf-D4 fragment contained the hSNF2H binding site based on coimmunoprecipitation and in vitro competition assays. As compared with other truncated mutants, expression of Rsf-D4 resulted in remarkable growth inhibition in ovarian cancer cells with Rsf-1 gene amplification and overexpression, but not in those without detectable Rsf-1 expression. The above findings suggest that interaction between Rsf-1 and hSNF2H may define a survival signal in those tumors overexpressing Rsf-1.
Article
Full-text available
New methods for the genome-wide analysis of chromatin are providing insight into its roles in development and their underlying mechanisms. Current studies indicate that chromatin is dynamic, with its structure and its histone modifications undergoing global changes during transitions in development and in response to extracellular cues. In addition to DNA methylation and histone modification, ATP-dependent enzymes that remodel chromatin are important controllers of chromatin structure and assembly, and are major contributors to the dynamic nature of chromatin. Evidence is emerging that these chromatin-remodelling enzymes have instructive and programmatic roles during development. Particularly intriguing are the findings that specialized assemblies of ATP-dependent remodellers are essential for establishing and maintaining pluripotent and multipotent states in cells.
Article
Full-text available
An estimated 25%-40% of infertile men have idiopathic infertility associated with deficient sperm numbers and quality. Here, we identify the membrane-anchored serine protease PRSS21, also known as testisin, to be a novel proteolytic factor that directs epididymal sperm cell maturation and sperm-fertilizing ability. PRSS21-deficient spermatozoa show decreased motility, angulated and curled tails, fragile necks, and dramatically increased susceptibility to decapitation. These defects reflect aberrant maturation during passage through the epididymis, because histological and electron microscopic structural analyses showed an increased tendency for curled and detached tails as spermatozoa transit from the corpus to the cauda epididymis. Cauda epididymal spermatozoa deficient in PRSS21 fail to mount a swelling response when exposed to hypotonic conditions, suggesting an impaired ability to respond to osmotic challenges facing maturing spermatozoa in the female reproductive tract. These data suggest that aberrant regulation of PRSS21 may underlie certain secondary male infertility syndromes, such as "easily decapitated" spermatozoa in humans.
Article
Full-text available
Centromeres are chromosomal structures required for equal DNA segregation to daughter cells, comprising specialized nucleosomes containing centromere protein A (CENP-A) histone, which provide the basis for centromeric chromatin assembly. Discovery of centromere protein components is progressing, but knowledge related to their establishment and maintenance remains limited. Previously, using anti-CENP-A native chromatin immunoprecipitation, we isolated the interphase-centromere complex (ICEN). Among ICEN components, subunits of the remodeling and spacing factor (RSF) complex, Rsf-1 and SNF2h proteins, were found. This paper describes the relationship of the RSF complex to centromere structure and function, demonstrating its requirement for maintenance of CENP-A at the centromeric core chromatin in HeLa cells. The RSF complex interacted with CENP-A chromatin in mid-G1. Rsf-1 depletion induced loss of centromeric CENP-A, and purified RSF complex reconstituted and spaced CENP-A nucleosomes in vitro. From these data, we propose the RSF complex as a new factor actively supporting the assembly of CENP-A chromatin.
Article
Full-text available
Author Summary While the chromatin of eukaryotes provides an efficient means to compact large amounts of DNA into a small nucleus, it renders the DNA relatively inaccessible. ATP-dependent chromatin remodeling complexes mobilize nucleosomes and provide a means to gain access to DNA in chromatin. While the biochemical functions of chromatin remodeling complexes is well-characterized, less is known of their biological functions. In this manuscript, we elucidate the biological functions of Bptf, a subunit of the NURF chromatin remodeling complex. Our studies show that Bptf is required for the establishment of the anterior–posterior axis of the mouse embryo during the earliest stages of development. To understand its functions in tissue differentiation, we generated and characterized Bptf-mutant ES cells. Mutant embryonic stem cells show significant defects in the differentiation of ectoderm, endoderm, and mesoderm. Genome-wide analysis of gene expression defects during differentiation has identified many Bptf-dependent pathways including key regulators of ectoderm, endoderm, and mesoderm differentiation. Moreover, we have identified critical functions for Bptf during the TGFβ/Smad-induced expression of visceral endoderm and mesoderm markers, an important signaling pathway in the early embryo. These results suggest that chromatin remodeling by Bptf regulates key signaling pathways in the early mouse embryo.
Article
The loss of Cecr2, which encodes a chromatin remodeling protein, has been associated with the neural tube defect (NTD) exencephaly and open eyelids in mice. Here, we show that two independent mutations of Cecr2 are also associated with specific inner ear defects. Homozygous mutant 18.5 days post coitus (dpc) fetuses exhibited smaller cochleae as well as rotational defects of sensory cells and extra cell rows in the inner ear reminiscent of planar cell polarity (PCP) mutants. Cecr2 was expressed in the neuroepithelium, head mesenchyme, and the cochlear floor. Although limited genetic interaction for NTDs was seen with Vangl2, a microarray analysis of PCP genes did not reveal a direct connection to this pathway. The mechanism of Cecr2 action in neurogenesis and inner ear development is likely complex.
Article
Over 200 mouse genes are associated with neural tube defects (NTDs), including Cecr2, the bromodomain-containing subunit of the CERF chromatin remodeling complex. Gene-trap mutation Cecr2(Gt45Bic) results in 74% exencephaly (equivalent of human anencephaly) on the BALB/c strain. Gene expression altered during cranial neural tube closure by the Cecr2 mutation was identified through microarray analysis of 11-14 somites stage Cecr2(Gt45Bic)embryos. Analysis of Affymetrix Mouse 430 2.0 chips detected 60 transcripts up-regulated and 54 transcripts down-regulated in the Cecr2(Gt45Bic) embryos (fold > 1.5, p < 0.05). The Cecr2 transcript was reduced only approximately 7- to 14-fold from normal levels, suggesting the Cecr2(Gt45Bic) is a hypomorphic mutation. We therefore generated a novel Cecr2 null allele (Cecr2 (tm1.1Hemc)). Resulting mutants displayed a stronger penetrance of exencephaly than Cecr2(Gt45Bic) in both BALB/c and FVB/N strains, in addition to midline facial clefts and forebrain encephalocele in the FVB/N strain. The Cecr2 transcript is reduced 260-fold in the Cecr2(tm1.1Hemc) line. Subsequent qRT-PCR using Cecr2 (tm1.1Hemc) mutant heads confirmed downregulation of transcription factors Alx1/Cart1, Dlx5, Eya1, and Six1. As both Alx1/Cart1 and Dlx5 mouse mutations result in exencephaly, we hypothesize that changes in expression of these mesenchymal/ectodermal transcription factors may contribute to NTDs associated with Cecr2.
Article
Following the discovery of mammalian sperm capacitation and its fundamental importance for the acquisition of fertilizing potential, it has gradually become possible to identify some specific molecules and molecular events that play pivotal roles in the "switching on" of spermatozoa. These are discussed in the context of the promotion and regulation of capacitation, emphasizing differences between commonly used conditions in vitro and the environment in vivo where spermatozoa normally undergo capacitation. Although typical culture media used in vitro do support capacitation, they do not prevent capacitated cells from undergoing spontaneous acrosome reactions and so losing fertilizing potential. This is not a problem in vitro, but could be in vivo where few spermatozoa reach the site of fertilization. Several small molecules, known to be present in vivo, have been shown in vitro to bind to spermatozoa and to regulate capacitation, first accelerating capacitation and then inhibiting spontaneous acrosome reactions, by regulating cAMP production. Since spermatozoa would contact these molecules during and after ejaculation, it is plausible that they serve a similar function in vivo. The mechanisms whereby the presence or absence of decapacitation factors might alter plasma membrane architecture and so alter functionality of a number of membrane-associated enzymes involved in capacitation are also considered. Finally, several unresolved issues relating to events during capacitation are discussed.