Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Molecular Cell, Vol.

6, 975–987, November, 2000, Copyright 2000 by Cell Press

The Mouse Spo11 Gene Is Required


for Meiotic Chromosome Synapsis

Peter J. Romanienko and R. Daniel Camerini-Otero* Hawley, 2000). For instance, meiotic recombination is
Genetics and Biochemistry Branch initiated by DNA DSBs generated by the action of the
National Institute of Diabetes and Digestive Spo11 protein (Spo11p) in S. cerevisiae (Keeney et al.,
and Kidney Diseases 1997), and in the absence of Spo11p, DSBs are not
National Institutes of Health formed and homologous chromosomes do not synapse
Bethesda, Maryland 20892 at wild-type levels (Giroux et al., 1989). Similarly, the
disruption of SPO11 homologs in S. pombe (Cervantes
et al., 2000), Drosophila (McKim et al., 1998), C. elegans
Summary (Dernburg et al., 1998), and Coprinus cinereus (Celerin
et al., 2000) results in defective gametes, and a reduction
The Spo11 protein initiates meiotic recombination by in meiotic recombination has been reported in S. cere-
generating DNA double-strand breaks (DSBs) and is visiae, S. pombe, Drosophila, and C. elegans. Chromo-
required for meiotic synapsis in S. cerevisiae. Surpris- somes still synapse at wild-type levels in spo11⫺/⫺ mu-
ingly, Spo11 homologs are dispensable for synapsis tants of the worm and fly but not in Coprinus cinereus.
in C. elegans and Drosophila yet required for meiotic In addition, meiosis is partially rescued by radiation-
recombination. Disruption of mouse Spo11 results in induced DSBs in budding yeast (Thorne and Byers,
infertility. Spermatocytes arrest prior to pachytene 1993; Gasior, 1999), worm (Dernburg et al., 1998), fly
with little or no synapsis and undergo apoptosis. We (R. Patel and K. McKim, personal communication), and
did not detect Rad51/Dmc1 foci in meiotic chromo- Coprinus cinereus (Celerin et al., 2000). These rescue
some spreads, indicating DSBs are not formed. Cis- experiments provide evidence for meiotic DSB forma-
platin-induced DSBs restored Rad51/Dmc1 foci and tion in eukaryotes other than S. cerevisiae and S. pombe
promoted synapsis. Spo11 localizes to discrete foci (Cervantes et al., 2000) and that the requirement for
during leptotene and to homologously synapsed chro- Spo11 can be partially bypassed. Thus, it appears that
mosomes. Other mouse mutants that arrest during in worm and fly, synapsis can be mediated by pairing
meiotic prophase (Atm ⫺/⫺, Dmc1 ⫺/⫺, mei1, and Morc⫺/⫺) centers that function independently of the need for mei-
showed altered Spo11 protein localization and expres- otic recombination. If dependence on pairing centers is
sion. We speculate that there is an additional role for seen with an increase in genome complexity, one might
Spo11, after it generates DSBs, in synapsis. easily imagine that mammals might also utilize pairing
centers.
Introduction Spo11-mediated DSBs occur at leptotene in S. cere-
visiae and disappear by zygotene (Roeder, 1997). This
Sexual reproduction and meiotic recombination are coincides with localization of Rad51 and Dmc1, two
common to most eukaryotes (Welch and Meselson, eukaryotic RecA homologs, to discrete foci in mouse
2000). Meiosis is a specialized cell division that reduces (Tarsounas et al., 1999) and in a Spo11-dependent man-
the normal chromosome complement by half in order ner in yeast (Bishop, 1994), the former presumably also
to generate haploid gametes. The proper segregation of at sites of DSBs. The presence of Rad51 and Dmc1 is
consistent with their biochemical function in DNA strand
chromosomes into gametes generally requires meiotic
exchange at the site of DNA DSBs.
recombination. In most organisms, genetic exchange
Spo11p is a member of a novel family of type II-like
occurs, and homologous chromosomes undergo synap-
topoisomerases, and consistent with that, site-directed
sis and form a synaptonemal complex (SC) (Heyting,
mutagenesis of a conserved tyrosine (Y135F) abolishes
1996). The SC is a proteinaceous structure composed
meiotic recombination (Bergerat et al., 1997). Homologs
of lateral elements (LE) and the central element (CE).
of Spo11p have been isolated from mouse, Spo11,
The LEs are parallel structures that originate as the axial
(Keeney et al., 1999; Romanienko and Camerini-Otero,
elements (AE), one component of which is the protein
1999; Shannon et al., 1999; Metzler-Guillemain and de
Scp3 (Dobson et al., 1994). The AEs are seen at leptotene
Massy, 2000) and man, SPO11, (Romanienko and Cam-
during meiotic prophase I and eventually form along erini-Otero, 1999; Shannon et al., 1999), and their ex-
the entire length of each homolog. Synapsis can be pression patterns are consistent with having a meiotic
visualized when two AEs of homologous chromosomes function. Using RT-PCR, many alternatively spliced vari-
come together during zygotene (Zickler and Kleckner, ants can be detected in testes, and Spo11 transcripts
1998). The CE is formed when transverse filaments (TFs), are also detected in somatic tissues. Analysis of Spo11
composed of the protein Scp1, bridge the axial elements expression in staged mouse spermatocytes showed the
during zygotene and pachytene (Schmekel and Dane- highest level of transcript during pachytene (Shannon
holt, 1998). et al., 1999), which is unexpected with Spo11 catalyzing
Whereas the initiation of meiotic recombination ap- DNA DSBs during leptotene. This would be more consis-
pears to be highly conserved, some eukaryotes differ tent with an additional role for Spo11 when homologous
in the molecular requirements for synapsis (Walker and chromosomes are fully synapsed. In fact, SPO11 tran-
script levels are also at their highest levels during pachy-
* To whom correspondence should be addressed (e-mail: camerini@ tene in yeast (Atcheson et al., 1987; Chu et al., 1998),
ncifcrf.gov). but no late function for Spo11p has been proposed.
Molecular Cell
976

Figure 1. Generation of Spo11-Deficient


Mice
(A) Gene disruption of the Spo11 locus. The
targeting construct and genomic locus of
Spo11 are shown. The EcoRI site in intron 1
was disrupted, and another EcoRI site was
created at the junction of the neo cassette
and exon 1 of Spo11 to facilitate identification
of targeted clones. The probe was derived
from the Spo11 cDNA encompassing exons
10–13. An EcoRI digest would give a 13 kb
band from wild-type genomic DNA and a 16
kb band from a targeted allele.
(B) Southern blot analysis of representative
genotypes of Spo11 knockout mice. Homo-
zygous knockout, ⫺/⫺; heterozygous, ⫹/⫺;
wild-type, ⫹/⫹. The digest was done with
EcoRI and probed with Spo11 cDNA, exons
10–13. The arrows indicate positions of bands
and their sizes are indicated.
(C) PCR genotyping of Spo11 knockout mice
by PCR. Mice used in experiments were ge-
notyped by PCR. The same mice were used
for Southern blot analysis in (B), indicating
the method is consistent with results from
Southern blotting. The primer locations are
indicated in (A). Targeting at the Spo11 locus
removes the binding site for one Spo11-spe-
cific primer.
(D) Northern analysis of Spo11 knockout
mice. Poly(A)⫹ RNA isolated from testes of
mice with the indicated genotypes was
probed with the mouse coding sequence
cDNA, and the same blot was later probed
with the mouse ␤-actin cDNA. The Spo11 transcript (1.8 kb) was absent in ⫺/⫺ testes and reduced in the heterozygote. The ␤-actin signal
is not relevant to the phenotype and serves as a loading standard. Postmeiotic ␣-actin was absent in ⫺/⫺ testes.

Here, we disrupted the mouse Spo11 gene and deter- offspring in a Mendelian fashion, and homozygotic
mined it is essential for gametogenesis and meiotic knockout mice did not exhibit any gross external mor-
chromosome synapsis. In addition, we found that Spo11 phological defects. Mice used for experiments were ge-
localizes to discrete foci early in meiosis and later to notyped by PCR on genomic DNA (Figure 1C). Northern
regions of homologous chromosome synapsis. Further- blot analysis of mRNA from adult testes from Spo11
more, Spo11 expression and localization of the protein ⫹/⫹, ⫹/⫺, and ⫺/⫺ mice (Figure 1D) showed that the
is altered in mutant mice that arrest during meiotic pro- transcript was reduced in heterozygotes and absent
phase I. Finally, we were able to partially rescue the from ⫺/⫺ testes. The same blot was probed with mouse
defect in synapsis in Spo11⫺/⫺ spermatocytes using the ␤-actin as a control for the loading of mRNA. Mouse
DSB-causing reagent cisplatin. ␤-actin cross-hybridizes to postmeiotically expressed
␣-actin (Hecht et al., 1984). The absence of ␣-actin in
Results Spo11⫺/⫺ testes is evidence that there is an arrest prior
to the appearance of postmeiotic cells. These results
Targeted Inactivation of Mouse Spo11 show that the Spo11 gene is inactivated in ⫺/⫺ mice.
The mouse Spo11 gene was inactivated by gene tar-
geting in embryonic stem (ES) cells. A targeting con- Spo11⫺/⫺ Male Mice Are Infertile
struct was made (Figure 1A) that removed part of the The first indication of a defect attributable to Spo11
coding sequence of exon 1 from 3 amino acids after the deficiency is the significant decrease in testicular size
second in-frame ATG up to nucleotide 35 of the first in mutant males (Figure 2A). The reduced size of the
intron (Keeney et al., 1999; Romanienko and Camerini- testis is similar to results seen in other mouse knockouts
Otero, 1999). The removal of most of exon 1 and part of early meiotic genes (Yoshida et al., 1998; Yuan et al.,
of intron 1 was designed to effectively create a protein 2000). Histological analysis of seminiferous tubule cross
null without greatly disturbing the genomic locus. Tar- sections from wild-type (Figure 2B) and mutant mice
geted clones were analyzed by Southern blotting, and a show many of the tubules in the mutants are barren,
correctly targeted cell line was injected into blastocysts. indicating massive cell death or nonproliferation, while
Southern blot analysis of representative wild-type, het- other tubules show clusters of cells morphologically
erozygotic, and double knockout mice (Figure 1B) is similar to zygotene spermatocytes (Figure 2C). There
shown. The ability to generate homozygous knockout were no cells with pachytene morphology in Spo11⫺/⫺
mice indicates Spo11 is not essential for mouse devel- tubules. Spermatocytes in Spo11⫺/⫺ mice underwent
opment. The mutated Spo11 allele was transmitted to apoptosis (Figure 2E), while few apoptotic cells were
Mouse Spo11 Is Required for Meiotic Synapsis
977

Figure 2. Histological Examination of Adult Testes from Spo11⫺/⫺ Mice


(A) Reduced testicular size in Spo11⫺/⫺ mice. Wild-type, ⫹/⫹; heterozygote, ⫹/⫺; and homozygote knockout, ⫺/⫺ testes were compared
from 6-week-old male sibs.
(B) Hematoxylin- and eosin-stained cross section of a seminiferous tubule from a wild-type adult mouse. Magnification is 400⫻.
(C) Hematoxylin- and eosin-stained cross sections of seminiferous tubules from a Spo11⫺/⫺ adult mouse. Magnification is 400⫻.
(D) Fluorescent TUNEL assay in wild-type mouse testis section; labeling was detected in some tubules near the basal lamina (not shown).
Magnification is 200⫻.
(E) Fluorescent TUNEL assay in Spo11⫺/⫺ mouse testis section; labeled cells (green fluorescence within tubules) were detected in about 20%
of tubules. Magnification is 200⫻; the arrow indicates positive tubule.

detected in wild-type mice (Figure 2D). Approximately pregnant Spo11⫺/⫺ mice after several months of breed-
20% of the tubules in the mutant had clusters of apo- ing with wild-type male mice.
ptotic cells, while wild-type mice only showed an occa-
sional apoptotic cell. Spermatocytes from Spo11⫺/⫺ Mice Arrest Prior
to Pachytene
Spo11⫺/⫺ Female Mice Are Infertile Spermatocytes from Spo11⫺/⫺ mice proceed normally
Histological cross sections of wild-type and mutant through leptotene (Figure 4A), and 40 centromeres are
adult ovaries showed a reduction in the number of folli- visualized by staining with CREST antisera, which labels
cles in the mutant (Figure 3A, wild type; Figure 3B, ⫺/⫺), centromeres (He and Brinkley, 1996). The presence of
indicating there is a severe phenotype in female mice 40 centromeres at this early stage indicates there is no
resulting in few or no mature oocytes. Defects can be disruption of sister chromatid cohesion at the centro-
seen as early as 15 dpc in the fetal ovaries of ⫺/⫺ mice meres in the absence of Spo11. Most spermatocytes
(Figure 3D) that are entering the early stages of meiosis arrest later at a zygotene-like stage (Figure 4B) where
I. In comparison with wild-type mice (Figure 3C), the there is little or no synapsis between chromosomes,
cells in the mutant fetal ovary look homogeneous. Cells whether homologous or nonhomologous. While the mei-
with partially condensed chromatin, indicating zygotene otic arrest is absolute, the failure to synapse is not.
cells in wild-type mice, were reduced in the absence of Approximately 5%–10% of the spread nuclei exhibit par-
Spo11 (Figure 3D). We were unsuccessful in obtaining tial, and in some instances, considerable synapsis (Fig-
Molecular Cell
978

Figure 3. Histological Examination of Adult and Fetal Ovaries from Spo11⫺/⫺ Mice
(A) Hematoxylin- and eosin-stained adult wild-type ovary. Arrow indicates maturing follicle. Eight to twelve follicles were generally seen per
section. Magnification is 200⫻.
(B) Hematoxylin- and eosin-stained adult Spo11⫺/⫺ ovary. Arrow indicates follicle, but note absence of any other follicles. No more than one
or two follicles were seen per section. Magnification is 200⫻.
(C) Hematoxylin- and eosin-stained wild-type fetal ovary, 15 dpc. The two female fetal mice were from the same dam. Arrows denote cells
with condensed chromatin. Magnification is 1000⫻.
(D) Hematoxylin- and eosin-stained Spo11⫺/⫺ fetal ovary, 15 dpc. Note fewer cells with condensed chromatin morphology as compared to
(C). The number of cells with this morphology was half of those seen in wild type. Magnification is 1000⫻.

ure 4G). The synapsis, indicated by Scp3 staining of Localization of Mouse Spo11 in Meiotic Chromosome
lateral elements, occurs mostly between nonhomologs Spreads from Spermatocytes
since frequent partner switches are observed. Scp3 Some meiotic proteins can be visualized with respect
staining of lateral elements colocalized with Scp1 stain- to meiotic chromosomes, and this approach often yields
ing (unpublished observations). insights into what role a specific protein may play during
Rad51 and Dmc1 are RecA-homologs, the latter meio- meiosis (Plug et al., 1998; Moens et al., 1999). Spo11
sis-specific, that are required for strand invasion (Shino- has not been visualized in meiotic nuclei of any organism
hara et al., 1997). Rad51 and Dmc1 are believed to load previously. We performed indirect immunofluorescence
onto single-stranded DNA generated at the sites of mei- of meiotic chromosome spreads using an affinity-puri-
otic DSBs. Neither Rad51 nor Dmc1 foci were seen in fied antibody raised against a peptide (MmPep-1) de-
Spo11⫺/⫺ spermatocytes (Figures 4D and 4F), but they rived from the Toprim domain (Aravind et al., 1998) of
were readily detected in wild-type spermatocytes (Fig- the mouse Spo11 protein. First, we performed several
ure 4C and 4E). The ␣Rad51 antibody routinely detected controls to make sure the antisera and purified antibody
one or two large signals in Spo11⫺/⫺ spermatocytes. were Spo11 specific. Preimmune sera did not recognize
These signals may be cellular reservoirs of Rad51 or SCs in meiotic spreads, but immune serum did. The
cross-reactivity to another protein. We do not believe signal from crude antiserum and purified antibody was
the latter is true, since we do not see this staining in abolished by incubation in the presence of 10-fold molar
wild-type nuclei. The absence of Rad51 and Dmc1 foci excess of purified MmPep-1 but not in the presence of
confirms that meiotic DSBs are not formed in the ab- 100-fold molar excess of an unrelated peptide. Rabbit
sence of Spo11. ␣-mouse Spo11 was able to detect the two major forms
Mouse Spo11 Is Required for Meiotic Synapsis
979

Figure 4. Indirect Immunofluorescence of Spo11⫺/⫺ Spermatocytes


(A) Leptotene stage Spo11⫺/⫺ spermatocyte. Merged image of ␣Scp3 (red) and the centromere antisera CREST (green). All images initially
viewed at 1000⫻ magnification. Scale bar, 10 ␮m. Overlap of red and green signals results in yellow signal.
(B) Zygotene-like stage of arrest in Spo11⫺/⫺ spermatocytes. Merged image of ␣Scp3 (red) and the centromere antisera CREST (green). Three
nuclei are shown.
(C) Zygotene wild-type spermatocyte; merged image of ␣Scp3 (red) and the ␣Rad51 (green).
(D) Zygotene-like Spo11⫺/⫺ spermatocyte; merged image of ␣Scp3 (red) and the ␣Rad51 (green).
(E) Zygotene wild-type spematocyte; merged image of ␣Scp3 (red) and the ␣Dmc1 (green).
(F) Zygotene-like Spo11⫺/⫺ spermatocyte; merged image of ␣Scp3 (red) and the ␣Dmc1 (green).
(G) Zygotene-like Spo11⫺/⫺ spematocyte showing extent of synapsis, indicated by arrows. Stained with ␣Scp3 (red).
Molecular Cell
980

Figure 5. Spo11 Localization in Meiotic Chromosome Spreads


(A) Spo11⫺/⫺ spermatocyte; merged image of ␣Scp3 (red) and the ␣Spo11 (green). Note absence of Spo11 signal. All images were initially
viewed at 1000⫻ magnification. Scale bar, 10 ␮m. Overlap of red and green signals results in yellow signal.
(B) Leptotene spermatocyte; merged image of ␣Scp3 (red) and the ␣Spo11 (green).
(C) Zygotene spermatocyte; merged image of ␣Scp3 (red) and the ␣Spo11 (green). Arrows indicate regions of synapsis.
(D) Pachytene spermatocyte; merged image of ␣Scp3 (red) and the ␣Spo11 (green). Arrow indicates the pseudoautosomal region (PAR).
(E) Magnification of (C); arrow indicates PAR.
(F) Pachytene spermatocyte; merged image of ␣Scp1 (red) and the ␣Spo11 (green). Arrow indicates PAR.
(G) Magnification of (E); arrow indicates PAR.
Mouse Spo11 Is Required for Meiotic Synapsis
981

Figure 6. Spo11 Localization and Expression in Mouse Meiotic Mutants


(A) RT-PCR analysis of Spo11 expression in testes of mutant mice. The two forms of Spo11 (␣ and ␤) are indicated by arrows in the top panel,
and the bottom panel shows control amplification of Aop2 (Pittman et al., 1998). The mutants analyzed are listed across the top. 13.5 dpp
(days postpartum), amplification from testis cDNA from a juvenile mouse; H20, water control in amplification reaction. ␣ and ␤ forms of Spo11
are shown with primer location relative to exon 2, active site region, and Toprim domain. The sizes of putative proteins are indicated on the
right. aa, amino acids.
(B) Localization of Spo11 in Dmc1⫺/⫺ spermatocyte. Merged image of ␣Scp3 (red) and ␣Spo11 (green). Arrowhead indicates Spo11 staining
in synapsed region, arrows indicate ends of axial elements of homologous chromosome participating in homologous synapsis, and asterisks
indicate synapsis between nonhomologs. Images were initially viewed at 1000⫻ magnification. The scale bar is 10 ␮m. Overlap of red and
green signals results in yellow signal.
(C) Localization of Spo11 in mei1 spermatocytes. Merged image of ␣Scp3 (red) and ␣Spo11 (green). Arrow indicates Spo11 staining of fully
synapsed homologs, and asterisks indicate synapsis between nonhomologs. Two nuclei are shown.

of Spo11 expressed from expression vectors trans- The next stage in meiosis is zygotene, during which
fected into mouse 3T3 cells but did not react with any homologous chromosomes begin to synapse (Zickler
other protein from untransfected 3T3 cells (data not and Kleckner, 1998). The distribution of Spo11 changes,
shown). The inability to visualize Spo11 foci or localiza- and localization is now to the regions of synapsis be-
tion at partially synapsed regions (Figure 5A) in Spo11⫺/⫺ tween homologs (Figure 5C). Full synapsis of homologs
mice is strong evidence that our antibody to Spo11 occurs during pachytene, and Spo11 decorates the
is specific. lengths of all autosomes (stained with ␣Scp3) in a dis-
We first detected Spo11 during the leptotene stage continuous pattern (Figure 5D). ␣Spo11 also stains the
of meiotic prophase I (Figure 5B). This is the stage when XY chromosome pair only at the pseudoautsomal region
meiotic DSBs are generated in budding yeast (Roeder, (PAR) (Figure 5D), where the X and Y chromosomes
1997). The majority of Spo11 foci (green) did not colocal- homologously synapse and recombine in the male (Sori-
ize with the antibody marker (␣Scp3, red) we used for ano et al., 1987). A magnification of this region (Figure
assessing the extent of axial element formation. These 5E) shows the PAR and the discontinuous localization
foci most likely represent sites of DNA DSB formation. of Spo11 in more detail. Spo11 colocalized with the
Molecular Cell
982

SC, as determined by Scp1 staining (Figure 5F). Similar Partial Rescue of Meiosis by the DNA Damaging
results are seen in a magnification of the PAR from Agent Cisplatin
Figure 5F (Figure 5G). Spo11 dissociates from the SC Cisplatin is a chemotherapeutic agent that forms many
during diplotene as homologs begin to separate. Before different adducts with DNA, removal of some of which
desynapsis is complete, all Spo11 localization with the would require formation of a DNA DSB (Bhattacharyya et
SC vanishes and staining becomes diffuse (data not al., 2000 and references therein). Previous work showed
shown). cisplatin increases the amount of meiotic crossover
products by nearly 2-fold (Hanneman et al., 1997). We
Spo11 Expression and Localization in Spermatocytes used cisplatin to induce DSBs in arrested spermato-
from Other Mutant Mice Arrested cytes in our Spo11⫺/⫺ mice to see whether they pro-
in Meiotic Prophase I gressed beyond the zygotene-like stage we routinely
In order to gain further insight into the roles Spo11 may observed. After cisplatin administration, mice were sac-
have in meiosis, we looked at Spo11 expression and rificed at different time points and analyzed. As shown
localization in several mutant backgrounds. Atm⫺/⫺ mice above, Spo11⫺/⫺ spermatocytes do not have Rad51 and
are deficient for the mouse homolog of the gene mutated Dmc1 foci (Figures 4D and 4F, respectively). Three days
in ataxia-telangiectasia (AT) patients that have a defect after cisplatin administration, we were able to detect
in DNA repair and are infertile (Barlow et al., 1996). Atm⫺/⫺ Rad51 (merged with Scp3, Figure 7A) and Dmc1 foci
spermatocytes also show mislocalization of Rad51 and (merged with Scp3, Figure 7B). Approximately 50% of
Dmc1 to chromatin and reduced localization of these the Rad51 and Dmc1 foci colocalized within nuclei (Fig-
proteins to the developing SC (Barlow et al., 1998). ure 7C). We did not see many nuclei with Rad51/Dmc1
Dmc1⫺/⫺ mice are mutant for the meiosis-specific mouse foci one day after cisplatin administration (data not
RecA homolog, Dmc1 (Pittman et al., 1998), and arrest shown), and at five days post cisplatin administration
but show normal localization of Rad51. The presence the results were much like those seen after three days
of Rad51 foci in both Atm⫺/⫺ and Dmc1⫺/⫺ spermatocytes (data not shown). Seven days after cisplatin administra-
is evidence that meiotic DSBs are made in these mu- tion, there were very few spermatocytes that stained
tants. mei1 mice were derived from mutagenized ES with ␣Scp3 or had Rad51 or Dmc1 foci; in fact, the
cells and are infertile (Munroe et al., 2000). Morc⫺/⫺ mice absolute number of cells isolated from the testis was
are the result of a random transgene insertion (Watson severely reduced (unpublished observations).
et al., 1998), and the mutated gene encodes a protein The extent of synapsis in cisplatin-treated mice was
with limited homology to GHL (GyraseB, Hsp90, Mut L) quantified by counting the number of centromeres in
ATPases (Inoue et al., 1999). 100 nuclei stained with CREST antisera in cisplatin-
In mouse and man there are two predominant, alterna- treated and untreated Spo11⫺/⫺ mice (Figure 7D). There
tively spliced forms of Spo11 (Romanienko and Camer- was a significant decrease in nuclei with 40 centromeres
ini-Otero, 1999; Shannon et al., 1999) that code for two (no synapsis) after cisplatin treatment and an increase
proteins of different sizes (unpublished data). RT-PCR of cells with evidence of considerable synapsis (31–33
analysis of Spo11 expression in a panel of mutant mice CREST foci). The true extent of synapsis was determined
revealed a common defect in generation of the predomi- by staining with ␣Scp1 and CREST antisera (Figure 7E).
nant spliced form of the transcript (Figure 6A). All mice Regions of synapsis usually extended away from centro-
showed a severe reduction in removal of exon 2 (Figure meres, and cells with fewer staining centromeres (c ⬍
6A, compare lanes 1–3 with lane 5). The splicing of an- b ⬍ a) had relatively more Scp1. Thus, cisplatin-gener-
other transcript, DNA ligase III, was analyzed in these ated DSBs promote synapsis in Spo11⫺/⫺ spermato-
mutant mice. Splicing of the ␤ form of DNA ligase III cytes. Generally, 30%–50% of nuclei showed consider-
occurs in pachytene spermatocytes (Mackey et al., able synapsis (as determined by ␣Scp1 staining) after
1997), but we saw no difference in the splicing pattern cisplatin administration, as compared to 5%–10% in
of this transcript in the mutant mice we analyzed (data untreated controls.
not shown).
The localization of mouse Spo11 was performed in Discussion
meiotic chromosome spreads from mouse mutants that
arrest during meiotic prophase I. The mouse mutants Spo11 is required for fertility in both male and female
used, for the most part, do not show complete homolo- mice, and the meiotic arrest we observed is similar to
gous synapsis. The distribution of Spo11 was different that seen in other mouse knockouts that cause infertility.
in Dmc1⫺/⫺ spermatocytes from that of wild-type mice. Spo11⫺/⫺ deficient spermatocytes do not undergo wild-
Spo11 only localized to a subset of synapsed regions, type levels of synapsis, and the synapsis that does occur
and the only consistent observation was that these re- is mostly between nonhomologs. The phenotype in
gions appeared to be homologously synapsed (Figure mouse is more similar to S. cerevisiae and C. cinereus
6B). In a mei1 mouse, there was considerably more than to C. elegans and Drosophila, where SC formation
synapsis, and the fluorescent intensity of Spo11 was just is essentially unaffected in Spo11 mutants. Thus, large
slightly lower than that seen in normal spermatocytes genome size and complexity alone do not imply a need
(Figure 6C). There appeared to be full synapsis between for a Spo11-independent mechanism for chromosome
homologs (arrow, Figure 6C) as determined by ␣Scp3 synapsis as seen in C. elegans and Drosophila. Our data
staining but there was also some nonhomologous syn- on the phenotype of this knockout is in agreement with
apsis as well (asterisks, Figure 6C). Morc⫺/⫺ mice un- that in the accompanying study of Baudat and cowork-
dergo very little synapsis, and Spo11 could not be de- ers (Baudat et al. 2000 [this issue of Molecular Cell]).
tected in those spermatocytes (data not shown). Spo11 was first localized to discrete foci during lepto-
Figure 7. Partial Rescue of Meiotic Arrest in Spo11⫺/⫺ Spermatocytes
(A) Induction of Rad51 foci in Spo11⫺/⫺ spermatocytes 3 days after cisplatin administration. Merged image of ␣Scp3 (red) and ␣Rad51 (green).
Cells were in zygotene-like stage. The images were initially viewed at 1000⫻ magnification. Four nuclei are shown; the scale bar is 10 ␮m.
Overlap of red and green signals results in yellow signal.
(B) Induction of Dmc1 foci in Spo11⫺/⫺ spermatocytes 3 days after cisplatin administration. Merged image of ␣Scp3 (red) and ␣Dmc1 (green).
Cells were in zygotene-like stage; six nuclei are shown.
(C) Colocalization of Rad51 and Dmc1 foci in Spo11⫺/⫺ spermatocyte 3 days after cisplatin administration. Merged image of ␣Rad51 (red) and
␣Dmc1 (green).
(D) Graphic representation of increased synapsis seen in Spo11⫺/⫺ spermatocytes after cisplatin administration. One ⫺/⫺ mouse was treated
with cisplatin for 3 days, the other was injected with saline. The mice were from the same litter and ⱖ5 weeks old when injected. One hundred
nuclei were analyzed for each sample, and the number of CREST signals (centromeres) were recorded.
(E) Colocalization of Scp1 and centromeres (CREST antisera) in Spo11⫺/⫺ spermatocytes 3 days after cisplatin administration. Merged image
of ␣Scp1 (red) and CREST (green). Three nuclei are shown, (a) has 33 centromeres; (b), 30, and (c), 28. In many instances, synapsis extends
away from the centromere. Note more ␣Scp1 staining coincident with fewer centromeres.
Molecular Cell
984

tene, when DSBs are believed to occur, and later only tene in wild-type mice (Figures 5B, 5C, and 5E). Spo11
along synapsed regions of homologous chromosomes makes DSBs during leptotene in yeast (Roeder, 1997),
during zygotene and pachytene, including the PAR of but the temporal events of meiotic recombination have
the XY bivalent. not been studied in great detail in mouse. The localiza-
The meiotic defect in Spo11⫺/⫺ mice is qualitatively tion of mouse Rad51 to discrete foci during leptotene
different from other mouse meiotic mutants that pre- (Tarsounas et al., 1999) is reasonable evidence that
sumably function downstream in the recombination DSBs occur at this stage. Our demonstration that the
process. For example, Atm⫺/⫺ and Dmc1⫺/⫺ spermato- formation of Rad51 foci in mouse is Spo11-dependent,
cytes also arrest during meiotic prophase I, but Atm just as in yeast, is further evidence that DSBs occur
plays a more global role in genome surveillance and during leptotene. Since we were able to detect Rad51
the effects are pleiotropic, while the meiotic arrest in foci in Spo11⫺/⫺ spermatocytes after administration of
Dmc1⫺/⫺ mice may be due to a checkpoint activated by cisplatin, it is therefore reasonable to conclude that
unrepaired DSBs. mouse Spo11 generates meiotic DSBs.
Our Spo11⫺/⫺ mice were not subjected to an intensive The discontinuous pattern of Spo11 on pachytene
analysis of somatic tissues to detect effects other than chromosomes (Figures 5C and 5E) is unlike that seen
a disruption of meiosis. Histological analysis of cross using antibodies against the lateral (Scp3) and central
sections from thymus, in which there is a high level (Scp1) element components of the SC, and it would be
of Spo11 expression in comparison to other somatic interesting to see whether the distribution of Spo11 is
tissues (Romanienko and Camerini-Otero, 1999), did not specific for each chromosome. That Spo11 is expressed
indicate any obvious alterations in the absence of Spo11 at its highest level in pachytene spermatocytes in mouse
(unpublished observations). The defect seen in fetal (Shannon et al., 1999) and during pachytene in S. cere-
ovary (Figures 3C and 3D) can be attributed to the mei- visiae (Atcheson et al., 1987; Chu et al., 1998) is consis-
otic function of Spo11, but it would be interesting to tent with its detection along synapsed regions of paired
look earlier in gonad development to see if the premei- homologs, but pachytene localization does not neces-
otic defect we see in fetal testes (data not shown) can sarily imply a role in pachytene.
be detected earlier in primordial germ cells. The morpho-
logical change we saw in Spo11⫺/⫺ fetal testes may be Mutant Mice Reveal a Spo11 Splicing Defect
a reflection of some defect in premeiotic S phase in in Arrested Spermatocytes and Suggest a Role
Spo11⫺/⫺ mice, as was seen in SPO11-deleted yeast for Spo11 in Stabilizing Homologous Synapsis
(Cha et al., 2000). Analysis of Spo11 localization and expression in several
mutant mouse lines allowed us to speculate on the alter-
Spo11 Is Required for Meiotic Chromosome Synapsis nate forms of Spo11 and what function they may encode.
Indirect immunofluorescent analysis of meiotic chromo- The RT-PCR results from the mutant mice show a link
some spreads from Spo11⫺/⫺ male mice showed that between meiotic progression from zygotene to pachy-
axial elements form but synapsis was severely affected tene and the removal of exon 2 from the Spo11 transcript
(Figure 4B). Almost all the synapsis that occurs is be- (Figure 6A). The predominant form of Spo11 in wild-type
tween nonhomologs, as revealed by frequent partner adult testes does not contain exon 2. The simplest model
switches. The lack of synapsis might be attributable to suggests that the larger form of Spo11 (Spo11␤) is the
the absence of meiotic DSBs, but it is also possible that catalytic form that generates DSBs, while the smaller
Spo11 plays a role in earlier homolog pairing (Weiner form (Spo11␣) plays a structural role in the developing
and Kleckner, 1994) and/or synapsis itself. Normal syn- SC. Further evidence for this model is the fact that Rad51
apsis in mouse is also dependent on a number of pro- foci (marker for DSBs) form in Dmc1⫺/⫺ spermatocytes
teins involved in meiotic recombination (Atm, Barlow et (Pittman et al., 1998) (where there is little of the smaller
al., 1998; Dmc1, Pittman et al., 1998; Yoshida et al., Spo11␣ transcript) with the same kinetics as wild-type
1998; Msh4, Kneitz et al., 2000; and Msh5, de Vries et mice. mei1 mice show more homologous synapsis than
al., 1999; Edelmann et al., 1999). Synapsis was also Dmc1 and Atm mutants (data presented above and un-
defective in Scp3⫺/⫺ males, but female mice were fertile published observations) and, coincidentally, more splic-
(Yuan et al., 2000). ing to Spo11␣. The finding that DNA ligase III-␤ splicing
The dependence of normal synapsis on recombina- is unaffected in the mutant mice we analyzed indicates
tion is central to the deficiency seen in Spo11⫺/⫺ sperma- there may be multiple checkpoints regulating different
tocytes. Either the failure to initiate recombination sig- facets of meiotic progression, and that the arrest per
nals an arrest and normal synapsis does not occur, or se is not responsible for the splicing defect.
the lack of recombination results in a subsequent failure Our interpretation of immunofluorescence results
to complete synapsis that triggers the arrest. A mutant from Dmc1⫺/⫺ and mei1 spermatocytes is that Spo11
mouse that can bypass the requirement for DSB forma- localizes to regions of synapsis between homologs. Very
tion and recombination and permits normal synapsis little staining is seen in regions of incorrect synapsis,
would argue for the second model. so one can speculate that Spo11 is required for synapsis
between homologs or perhaps stabilizes synapsis be-
Spo11 Localization Is Consistent with Roles tween homologs. It is not clear whether Spo11 can dis-
in DSB Formation and Synapsis criminate between homologous and nonhomologous
Spo11 was localized in discrete foci during leptotene synapsis or whether the SC formed between nonhomo-
(Figure 5A) and more intensely at synapsed regions of logs is fundamentally different in a way that is not imme-
homologous chromosomes during zygotene and pachy- diately obvious with our cytological criteria.
Mouse Spo11 Is Required for Meiotic Synapsis
985

We note, however, that this picture might be an over- and the Micro Poly(A) Pure Kit (Ambion). Two micrograms of purified
simplification. Preliminary results indicate that Brca1Co/Co mRNA was run, transferred, and probed sequentially with the mouse
Spo11 cDNA and the mouse ␤-actin cDNA (Sigma). Tissues for
mice can progress and arrest in pachytene with fully
histological examination were removed and fixed overnight in neu-
synapsed chromosomes but still do not splice out exon tral-buffered 10% formalin (Sigma). Tissue was embedded in paraf-
2 (unpublished observations). fin and 6 ␮m sections were cut. Fetal mice were removed from
the mother and placed in fixative. The fetal age of the mice was
Partial Meiotic Rescue in Spo11⫺/⫺ Spermatocytes determined by comparison of external facial and body morphology,
by Cisplatin Argues for a Role of Spo11 and their age was placed at 15 dpc (Rugh, 1968). Before fixation, a
piece of tail was removed for genotyping. Male mice were confirmed
in Generating DSBs
by PCR amplification of the mouse male-specific Sry gene (GenBank
We partially rescued the meiotic arrest seen in our Spo11 accession number U70641) using primers Sry5⬘, 5⬘-GCCCAGCA
knockout by generating DNA damage with the chemo- GAATCCCAGCATGCA-3⬘ and Sry3⬘, 5⬘-TTTTGTTGAGGCAACTG
therapeutic agent cisplatin. We were able to detect CAGGCTG-3⬘. The PCR product was 250 bp, and the cycling proto-
Rad51/Dmc1 foci, the presence of which is good evi- col was 94⬚C, 2 min; 94⬚C, 15 s; and 68⬚C, 45 s for 35 cycles followed
dence for formation of cisplatin-induced DSBs. We were by 72⬚C, 10 min. Sections were stained with hematoxylin and eosin
also able to promote synapsis, indicating that DNA DSBs (H&E) or analyzed for apoptosis using the TACS 2 TdT-In situ Apo-
ptosis Detection Kit (Trevigen).
may influence SC formation, but wild-type levels of syn-
apsis were not observed. In any case, DSBs of any
Antibodies for Immunofluorescence
origin, whether generated by Spo11 or cisplatin, and
An antibody was raised against a peptide, MmPep-1 (amino acids
recombination are necessary and sufficient to promote 224–242, EKDATFQRLLDDNFCSRMS, encoded in exon 8 of mouse
significant levels of synapsis, and the postulated second Spo11 [Romanienko and Camerini-Otero, 1999]), that was synthe-
role of Spo11 may be in stabilizing homologous syn- sized on MAP (Multi-Antigenic Peptide) resin (Novabiochem) on a
apsis. PerSeptive Biosystems peptide synthesizer. The crude synthesis
product was used to generate polyclonal antisera in two rabbits
Experimental Procedures (Covance). Antisera was checked by Western blot analysis of mouse
Spo11:GFP fusion protein expressed in mouse 3T3 cells (ATCC).
Targeted Inactivation of the Mouse Spo11 Gene ␣-Spo11 was affinity purified by passage over a column to which
The targeting construct was based on ploxPneo (Yang et al., 1998) was coupled HPLC-purified MmPep-1. Peptide coupling was done
(a gift from Chuxia Deng). A 3.3 kb BspEI/ApaI genomic fragment using AminoLink Coupling Gel (Pierce), and binding and elution
containing the 5⬘ region of mouse Spo11 was blunted and cloned were done with ImmunoPure Gentle Binding buffer and ImmunoPure
into the blunted EcoRI site in ploxPneo between the TK gene and Gentle Elution buffer, respectively (Pierce). The purified antiserum
PGKneo cassette in the opposite orientation of transcription of the was dialyzed into Tris-buffered saline, .05% sodium azide, and con-
neo gene. This construct was designated pTarget1. A 1.1 kb SmaI/ centrated to approximately 1mg/ml using Slide-A-Lyzer Concentrat-
EcoRV genomic fragment containing intron 1 sequence was cloned ing Solution (Pierce). The purified antibody was aliquoted and frozen
into XhoI-cut, blunted pTarget1 on the other side of the neo cassette. at ⫺20⬚C.
This step removed the first 35 nucleotides of intron 1, which con- Primary antibodies (in addition to rabbit ␣-mouse Spo11 at 1:10
tained the splice donor site. This effectively deleted 140 bp of the or 1:20) used for immunofluorescence were mouse ␣-rat SCP1 at
Spo11 locus and inserted the neo cassette; the construct was desig- 1:10 (a gift from Christa Heyting, Wageningen University, The Neth-
nated pTarget2. A 9 kb EcoRI/AatII genomic fragment containing erlands), mouse ␣-hamster SCP3 (Cor1) at 1:1000 (a gift from Peter
exons 2–9 was blunted and cloned into pTarget2 cut with Hpa I, Moens, York University, Toronto, Canada), CREST human antisera
resulting in the final 20 kb targeting vector, pTarget3. The final clon- at 1:200 (a gift from Bill Brinkley, Baylor College, Texas), goat
ing step resulted in the deletion of an additional 110 bp from intron ␣-Dmc1 at 1:200 (C-20) (Santa Cruz Biotechnology), and rabbit
1, including the EcoRI site. PTarget3 was linearized with NotI and ␣-Rad51 at 1:200 (H-92) (Santa Cruz Biotechnology). All secondary
prepared for targeting. Ninety micrograms of linearized pTarget3 antibodies (Jackson Immunoresearch Laboratories) were used at
was electroporated into 1.8 ⫻ 107 TC1 ES cells (Deng et al., 1996) 1:200. Secondary antibodies were either conjugated with Rhoda-
(a gift from Chu-Xia Deng) and seeded onto mitomycin C–treated mine Red-X or FITC and were generated in goats or donkeys.
mouse embryonic feeder cells (Incyte Genomics). Colonies were
selected in media containing LIF (GIBCO–BRL), 350 ␮g/ml G418,
Meiotic Chromosome Spread Preparation
and 5 ␮M gancyclovir (a gift from Rick Proia). Colonies were picked,
and Immunofluorescence
expanded, and genomic DNA was extracted for Southern analysis.
Chromosome spreads were prepared as follows: testes were dis-
The removal of the EcoRI site in intron 1 and insertion of the neo
sected, seminiferous tubules were digested with .5 mg/ml collage-
cassette made the mutated allele 3 kb (16 kb) larger than the wild-
nase in RPMI 1640 high glucose media supplemented with amino
type allele (13 kb). The probe was derived from exons 10–13 of the
mouse Spo11 cDNA. One correctly targeted ES clone was injected acids (GIBCO–BRL) for 20 min at 33⬚C with gentle shaking, digested
into C57/B6 blastocysts to obtain germline transmission. Mice ob- with .25 mg/ml trypsin, 1 ␮g/ml DNase I for 20 min at 33⬚C with
tained after germline transmission were screened by PCR on geno- gentle shaking, then filtered through a 40 ␮m cell strainer (Falcon).
mic DNA isolated from tail tips. Primers for PCR genotyping were Soybean trypsin inhibitor, SBTI (GIBCO–BRL), was added at 100
Spo11-1, 5⬘-TGTCCCGCGGTCAGTGGTGCAG-3⬘ and Spo11-2, ␮g/ml, and the cell suspension was centrifuged at 800 ⫻ g for 5
5⬘-TCCAGGGCGTCGAAGAACGAGG-3⬘, which amplified a 150 bp min, washed with media containing SBTI, and recentrifuged. The
product from genomic DNA. Spo11-2 lies within the deleted region supernatant was removed, and cells were resuspended in .5% NaCl
and would not be present in the targeted allele. The neo primers were along with any residual media that remained after aspiration. The
Neo5⬘-Target, 5⬘-GTACTCGGATGGAAGCCGGTCTT-3⬘ and Neo3⬘- cell suspension was pipetted onto glass slides with hydrophobic
Target, 5⬘-GCCAAGCTCTTCAGCAATATCACG-3⬘. The amplified rings (Becton Dickinson), and the cells were allowed to swell. When
product was 280 bp and was only present if the targeted allele was the cells began to adhere to the slide surface, the slides were fixed
present. The PCR protocol was 94⬚C, 2 min; 94⬚C, 15 s; and 68⬚C, in 2% paraformaldehyde, .03% SDS for 3 min, then in 2% parafor-
45 s for 32 cycles followed by 72⬚C, 10 min with 100 ng of genomic maldehyde for 3 min. Slides were washed 3⫻ in .4% Photo-Flo 200
DNA in a 20 ␮l reaction using the Advantage cDNA Polymerase Mix (Kodak) and allowed to air dry.
(Clontech). Slides were washed 2⫻ in PBS, blocking solution (10% serum
from goat or donkey, 3% BSA, .05% Triton X-100 in PBS) was added
Analysis of Spo11 Knockout Mice to the hydrophobic rings, and slides were incubated for 2 hr in a
Northern blot analysis was done on poly(A)⫹ RNA prepared from humid chamber at room temperature. Primary antibodies were di-
testes of the corresponding genotypes using Trizol (GIBCO–BRL) luted in blocking buffer, and slides were incubated in a humid cham-
Molecular Cell
986

ber at 37⬚C for 1 hr. After washing, secondary antibodies were added Barlow, C., Hirotsune, S., Paylor, R., Liyanage, M., Eckhaus, M.,
for 20 min at room temperature. The slides were washed and allowed Collins, F., Shiloh, Y., Crawley, J.N., Ried, T., Tagle, D., and Wyn-
to dry. Vectashield Mounting Medium with DAPI (Vector Labora- shaw-Boris, A. (1996). Atm-deficient mice: a paradigm of ataxia
tories) was added and the slides were viewed. The chromosome telangiectasia. Cell 86, 159–171.
spread and antibody incubation procedure was adapted from a Barlow, C., Liyanage, M., Moens, P.B., Tarsounas, M., Nagashima,
protocol from Peter Moens. K., Brown, K., Rottinghaus, S., Jackson, S.P., Tagle, D., Ried, T., and
Slides were examined with a Leica fluorescent microscope, and Wynshaw-Boris, A. (1998). Atm deficiency results in severe meiotic
individual FITC and rhodamine images were captured as eight bit disruption as early as leptonema of prophase I. Development 125,
color images at 1000⫻ magnification using an oil immersion lens. 4007–4017.
The images were processed using Adobe Photoshop 4.0 and
Baudat, F., Manova, K., Yuen, J.P., Jasin, M., and Keeney, S. (2000).
merged.
Chromosome synapsis and sexually dimorphic meiotic progression
Balb/c mice were used for localization of Spo11. Juvenile male
in mice lacking Spo11. Mol. Cell 6, this issue, 989–998.
mice of ages 10–21 days postpartum (dpp) were used to prepare
the spreads. The mutant mice used in this study are as follows, Bergerat, A., de Massy, B., Gadelle, D., Varoutas, P.C., Nicolas, A.,
Atm⫺/⫺ (Barlow et al., 1996) (purchased from Jackson Laboratories), and Forterre, P. (1997). An atypical topoisomerase II from Archaea
Dmc1⫺/⫺ (Pittman et al., 1998) and mei1 (Munroe et al., 2000) (a gift with implications for meiotic recombination. Nature 386, 414–417.
from John Schimenti, Jackson Laboratories, Bar Harbor, Maine), Bhattacharyya, A., Ear, U.S., Koller, B.H., Weichselbaum, R.R., and
and Morc⫺/⫺ (Watson et al., 1998) (a gift from Andrew Zinn and Bishop, D.K. (2000). The breast cancer susceptibility gene BRCA1
Mark Watson, University of Texas-Southwestern, Dallas, Texas). The is required for subnuclear assembly of Rad51 and survival following
mutant mice were ⱖ5 weeks of age at time of assaying. treatment with the DNA cross-linking agent cisplatin. J. Biol. Chem.
275, 23899–23903.
Comparison of Spo11 Transcripts in Mutant and Wild-Type Bishop, D.K. (1994). RecA homologs Dmc1 and Rad51 interact to
Mice by RT-PCR form multiple nuclear complexes prior to meiotic chromosome syn-
The primers used to amplify mouse Spo11 were 5⬘MmSpo, 5⬘-CTG apsis. Cell 79, 1081–1092.
TTGGCCATGGTGAGAGAGG-3⬘ and MmSpoSeq1, 5⬘-TCCTTGAA Celerin, M., Merino, S.T., Stone, J.E., Menzie, A.M., and Zolan, M.E.
TGTTAGTCGGCACAGC-3⬘. The product with exon 2 is 560 bp and
(2000). Multiple roles of Spo11 in meiotic chromosome behavior.
without exon 2 is 460 bp. The PCR protocol was 94⬚C, 2 min; 94⬚C,
EMBO J. 19, 2739–2750.
15 s; and 68⬚C, 1 min for 33 cycles followed by 72⬚C, 10 min. The
Aop2 control PCR and primers were as described (Pittman et al., Cervantes, M.D., Farah, J.A., and Smith, G.R. (2000). Meiotic DNA
1998). Poly(A)⫹ RNA was isolated from mouse testis tissue as de- breaks associated with recombination in S. pombe. Mol. Cell 5,
scribed earlier, and cDNA synthesis was done as described (Ro- 883–888.
manienko and Camerini-Otero, 1999). Cha, R.S., Weiner, B.M., Keeney, S., Dekker, J., and Kleckner, N.
(2000). Progression of meiotic DNA replication is modulated by inter-
Cisplatin Administration chromosomal interaction proteins, negatively by Spo11p and posi-
Cisplatin (Sigma) was administered as described (Hanneman et al., tively by Rec8p. Genes Dev. 14, 493–503.
1997). Mice were sacrificed at time points indicated, and testes Chu, S., DeRisi, J., Eisen, M., Mulholland, J., Botstein, D., Brown,
were removed for meiotic chromosome spread preparation and/or P.O., and Herskowitz, I. (1998). The transcriptional program of sporu-
histological examination. CREST foci were counted in a methodical lation in budding yeast. Science 282, 699–705.
manner with no bias in nuclei selection other than the omission of de Vries, S.S., Baart, E.B., Dekker, M., Siezen, A., de Rooij, D.G., de
leptotene spermatocytes, which are apparently normal in Spo11⫺/⫺ Boer, P., and te Riele, H. (1999). Mouse MutS-like protein Msh5 is
mice. All cells were in a zygotene-like stage. Foci that were partially required for proper chromosome synapsis in male and female meio-
overlapped such that there was no space between them were sis. Genes Dev. 13, 523–531.
counted as one focus. Adminsitration of cisplatin was performed
Deng, C., Wynshaw-Boris, A., Zhou, F., Kuo, A., and Leder, P. (1996).
according to ACUC guidelines at the NIH.
Fibroblast growth factor receptor 3 is a negative regulator of bone
growth. Cell 84, 911–921.
Acknowledgments
Dernburg, A.F., McDonald, K., Moulder, G., Barstead, R., Dresser,
We are grateful to Chu-Xia Deng and Cuiling Li for blastocyst injec- M., and Villeneuve, A.M. (1998). Meiotic recombination in C. elegans
tion, generating chimeric mice, and providing ES cells; Rick Proia initiates by a conserved mechanism and is dispensable for homolo-
and his lab for the gancyclovir and targeting advice; April Anderson gous chromosome synapsis. Cell 94, 387–398.
for technical assistance; Linda Robinson for her assistance; and Dobson, M.J., Pearlman, R.E., Karaiskakis, A., Spyropoulos, B., and
George Poy for oligo and peptide synthesis. We would like to thank Moens, P.B. (1994). Synaptonemal complex proteins: occurrence,
those who graciously provided the mutant mice, John Schimenti for epitope mapping and chromosome disjunction. J. Cell Sci. 107,
the Dmc1⫺/⫺ and mei mice, and Mark Watson and Andrew Zinn for 2749–2760.
the Morc⫺/⫺ mice. We are especially grateful to Bill Brinkley for the Edelmann, W., Cohen, P.E., Kneitz, B., Winand, N., Lia, M., Heyer, J.,
␣CREST antisera, Christa Heyting for the ␣SCP1 antibody, and Peter Kolodner, R., Pollard, J.W., and Kucherlapati, R. (1999). Mammalian
Moens for the ␣SCP3 antisera and advice on performing meiotic MutS homologue 5 is required for chromosome pairing in meiosis.
spreads. Finally, we want to thank Michael Lichten and Rick Proia Nat. Genet. 21, 123–127.
for their insightful comments on the manuscript and Chu-Xia Deng
Gasior, S.L. (1999). Assembly of recombination complexes in Sac-
and Kim McKim for communicating unpublished results.
charomyces cerevisiae. In Molecular Genetics and Cell Biology (Chi-
cago: University of Chicago), pp. 239.
Received August 4, 2000; revised September 19, 2000.
Giroux, C.N., Dresser, M.E., and Tiano, H.F. (1989). Genetic control
of chromosome synapsis in yeast meiosis. Genome 31, 88–94.
References
Hanneman, W.H., Legare, M.E., Sweeney, S., and Schimenti, J.C.
Aravind, L., Leipe, D.D., and Koonin, E.V. (1998). Toprim- a con- (1997). Cisplatin increases meiotic crossing-over in mice. Proc. Natl.
served catalytic domain in type IA and II topoisomerases, DnaG- Acad. Sci. USA 94, 8681–8685.
type primases, OLD family nucleases and RecR proteins. Nucleic He, D., and Brinkley, B.R. (1996). Structure and dynamic organization
Acids Res. 26, 4205–4213. of centromeres/prekinetochores in the nucleus of mammalian cells.
Atcheson, C.L., DiDomenico, B., Frackman, S., Esposito, R.E., and J. Cell Sci. 109, 2693–2704.
Elder, R.T. (1987). Isolation, DNA sequence, and regulation of a Hecht, N.B., Kleene, K.C., Distel, R.J., and Silver, L.M. (1984). The
meiosis-specific eukaryotic recombination gene. Proc. Natl. Acad. differential expression of the actins and tubulins during spermato-
Sci. USA 84, 8035–8039. genesis in the mouse. Exp. Cell Res. 153, 275–280.
Mouse Spo11 Is Required for Meiotic Synapsis
987

Heyting, C. (1996). Synaptonemal complexes: structure and func- Thorne, L.W., and Byers, B. (1993). Stage-specific effects of X-irradi-
tion. Curr. Opin. Cell Biol. 8, 389–396. ation on yeast meiosis. Genetics 134, 29–42.
Inoue, N., Hess, K.D., Moreadith, R.W., Richardson, L.L., Handel, Walker, M.Y., and Hawley, R.S. (2000). Hanging on to your homolog:
M.A., Watson, M.L., and Zinn, A.R. (1999). New gene family defined the roles of pairing, synapsis and recombination in the maintenance
by MORC, a nuclear protein required for mouse spermatogenesis. of homolog adhesion. Chromosoma 109, 3–9.
Hum. Mol. Genet. 8, 1201–1207. Watson, M.L., Zinn, A.R., Inoue, N., Hess, K.D., Cobb, J., Handel,
Keeney, S., Giroux, C.N., and Kleckner, N. (1997). Meiosis-specific M.A., Halaban, R., Duchene, C.C., Albright, G.M., and Moreadith,
DNA double-strand breaks are catalyzed by Spo11, a member of a R.W. (1998). Identification of morc (microrchidia), a mutation that
widely conserved protein family. Cell 88, 375–384. results in arrest of spermatogenesis at an early meiotic stage in the
Keeney, S., Baudat, F., Angeles, M., Zhou, Z.H., Copeland, N.G., mouse. Proc. Natl. Acad. Sci. USA 95, 14361–14366.
Jenkins, N.A., Manova, K., and Jasin, M. (1999). A mouse homolog Weiner, B.M., and Kleckner, N. (1994). Chromosome pairing via mul-
of the Saccharomyces cerevisiae meiotic recombination DNA trans- tiple interstitial interactions before and during meiosis in yeast. Cell
esterase Spo11p. Genomics 61, 170–182. 77, 911–991.
Kneitz, B., Cohen, P.E., Avdievich, E., Zhu, L., Kane, M.F., Hou, H., Welch, D.M., and Meselson, M. (2000). Evidence for the evolution
Kolodner, R.D., Kucherlapati, R., Pollard, J.W., and Edelmann, W. of bdelloid rotifers without sexual reproduction or genetic exchange.
(2000). MutS homolog 4 localization to meiotic chromosomes is Science 288, 1211–1215.
required for chromosome pairing during meiosis in male and female Yang, X., Li, C., Xu, X., and Deng, C. (1998). The tumor suppressor
mice. Genes Dev. 14, 1085–1097. SMAD4/DPC4 is essential for epiblast proliferation and mesoderm
Mackey, Z.B., Ramos, W., Levin, D.S., Walter, C.A., McCarrey, J.R., induction in mice. Proc. Natl. Acad. Sci. USA 95, 3667–3672.
and Tomkinson, A.E. (1997). An alternative splicing event which Yoshida, K., Kondoh, G., Matsuda, Y., Habu, T., Nishimune, Y., and
occurs in mouse pachytene spermatocytes generates a form of DNA Morita, T. (1998). The mouse RecA-like gene Dmc1 is required for
ligase III with distinct biochemical properties that may function in homologous chromosome synapsis during meiosis. Mol. Cell 1,
meiotic recombination. Mol. Cell. Biol. 17, 989–998. 707–718.
McKim, K.S., Green-Marroquin, B.L., Sekelsky, J.J., Chin, G., Yuan, L., Liu, J.G., Zhao, J., Brundell, E., Daneholt, B., and Hoog, C.
Steinberg, C., Khodosh, R., and Hawley, R.S. (1998). Meiotic synap- (2000). The murine Scp3 gene is required for synaptonemal complex
sis in the absence of recombination. Science 279, 876–878. assembly, chromosome synapsis, and male fertility. Mol. Cell 5,
Metzler-Guillemain, C., and de Massy, B. (2000). Identification and 73–83.
characterization of an SPO11 homolog in the mouse. Chromosoma Zickler, D., and Kleckner, N. (1998). The leptotene-zygotene transi-
109, 133–138. tion of meiosis. Annu. Rev. Genet. 32, 619–697.
Moens, P.B., Tarsounas, M., Morita, T., Habu, T., Rottinghaus, S.T.,
Freire, R., Jackson, S.P., Barlow, C., and Wynshaw-Boris, A. (1999).
The association of ATR protein with mouse meiotic chromosome
cores. Chromosoma 108, 95–102.
Munroe, R.J., Bergstrom, R.A., Zheng, Q.Y., Libby, B., Smith, R.,
John, S.W., Schimenti, K.J., Browning, V.L., and Schimenti, J.C.
(2000). Mouse mutants from chemically mutagenized embryonic
stem cells. Nat. Genet. 24, 318–321.
Pittman, D.L., Cobb, J., Schimenti, K.J., Wilson, L.A., Cooper, D.M.,
Brignull, E., Handel, M.A., and Schimenti, J.C. (1998). Meiotic pro-
phase arrest with failure of chromosome synapsis in mice deficient
for Dmc1, a germline-specific RecA homolog. Mol. Cell 1, 697–705.
Plug, A.W., Peters, A.H., Keegan, K.S., Hoekstra, M.F., de Boer, P.,
and Ashley, T. (1998). Changes in protein composition of meiotic
nodules during mammalian meiosis. J. Cell Sci. 111, 413–423.
Roeder, G.S. (1997). Meiotic chromosomes: it takes two to tango.
Genes Dev. 11, 2600–2621.
Romanienko, P.J., and Camerini-Otero, R.D. (1999). Cloning, charac-
terization, and localization of mouse and human SPO11. Genomics
61, 156–169.
Rugh, Roberts (1968). The Mouse (New York: Oxford University
Press).
Schmekel, K., and Daneholt, B. (1998). Evidence for close contact
between recombination nodules and the central element of the syn-
aptonemal complex. Chromosome Res. 6, 155–159.
Shannon, M., Richardson, L., Christian, A., Handel, M.A., and Thelen,
M.P. (1999). Differential gene expression of mammalian SPO11/
TOP6A homologs during meiosis. FEBS Lett. 462, 329–334.
Shinohara, A., Gasior, S., Ogawa, T., Kleckner, N., and Bishop, D.K.
(1997). Saccharomyces cerevisiae recA homologs RAD51 and
DMC1 have both distinct and overlapping roles in meiotic recombi-
nation. Genes Cells 2, 615–629.
Soriano, P., Keitges, E.A., Schorderet, D.F., Harbers, K., Gartler,
S.M., and Jaenisch, R. (1987). High rate of recombination and double
crossovers in the mouse pseudoautosomal region during male mei-
osis. Proc. Natl. Acad. Sci. USA 84, 7218–7220.
Tarsounas, M., Morita, T., Pearlman, R.E., and Moens, P.B. (1999).
RAD51 and DMC1 form mixed complexes associated with mouse
meiotic chromosome cores and synaptonemal complexes. J. Cell
Biol. 147, 207–219.

You might also like