You are on page 1of 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/229686244

Meiosis

Chapter · September 2010


DOI: 10.1002/9780470015902.a0005772.pub2

CITATION READS

1 17,741

1 author:

Maj Anita Hulten


Karolinska Institutet, Solna, Sweden
284 PUBLICATIONS 10,478 CITATIONS

SEE PROFILE

All content following this page was uploaded by Maj Anita Hulten on 27 February 2018.

The user has requested enhancement of the downloaded file.


Meiosis Advanced article

Maj A Hultén, University of Warwick, Coventry, UK Article Contents


. Introduction
Based in part on the previous version of this Encyclopedia of Life Sciences . Chromosome Pairing of Homologous Parental
(ELS) article, Meiosis by Charles Tease and Maj A Hultén. Chromosomes at Meiosis I

. Tools for Identification of Meiosis-specific Chromosome


Structures
. An Integrated Molecular and Cytogenetic Model of the
Crossing-over Process

. Chromosome Segregation

. Consequences of Abnormal Meiosis

Online posting date: 15th September 2010

Meiosis is a specialised type of cell division, the principal meiosis occurs during prophase of the first meiotic division
function of which is to produce spores/gametes (sperm (prophase I). Prophase I is conventionally subdivided into
and eggs in mammals) that have the haploid number of five stages (leptotene, zygotene, pachytene, diplotene and
chromosomes. In humans, this represents a reduction diakinesis) that reflect variation in chromosome organisa-
from 46 (23 pairs) to 23 chromosomes (one complete set) tion and behaviour during this period (Table 1).
Meiosis I has three crucial features that distinguish it
in sperm and eggs. The normal somatic number of 46
from mitosis: homologous chromosome pairing, reciprocal
chromosomes is restored at fertilisation. The most com-
recombination (also called crossing-over and chiasma
plex part of meiosis (prophase I) involves intimate pairing formation) and importantly a two-step release of sister
and synapsis of the homologous chromosomes followed chromatid cohesion that allows the segregation of the
by reciprocal recombination/crossing-over/chiasma for- homologues on the first meiotic spindle, followed by the
mation. In most organisms, chiasma formation is obliga- separation of sister chromatids during meiosis II. For ease
tory to allow the regular segregation of the (rearranged) of description, these aspects of chromosome behaviour are
homologues at the first (reductional) division (anaphase presented separately. However, it is important to remem-
I). In mammals, there are substantial differences between ber that they are interdependent events. Thus, the process
the two sexes as regards the meiotic process. Mammalian of chromosome pairing, reciprocal recombination/cross-
female prophase I takes place during foetal development, ing-over/chiasma formation are very intimately connected,
and the second meiotic cell division is not completed until and correct segregation of homologous chromosomes at
the first meiotic division can only be accomplished after
after fertilisation. In sharp contrast, mammalian male
successful recombinatorial processes having taken place
meiosis does not start until puberty and is continuous
beforehand, that is, at meiosis I prophase.
throughout life. Completion of reciprocal recombination/crossing-over
between parental half chromosomes (chromatids), toge-
ther with sister chromatid cohesion, leads to the formation
of chiasmata. Chiasmata bind homologous chromosomes
physically together to allow the chromosome pair (biva-
lent) to orientate at metaphase I such that the chromosome
Introduction movement centres (kinetochores located at the centro-
meres) are facing opposite spindle poles. This mech-
Meiosis consists of one round of deoxyribonucleic acid anistically favourable position is of vital importance for
(DNA) replication followed by two nuclear divisions. The proper kinetochore attachment to opposite spindles, pro-
two divisions (meiosis I and II) are divided into the same moting regular chromosome segregation at the subsequent
stages (prophase, metaphase, anaphase and telophase) as anaphase I (Figure 1).
mitosis (Table 1 and Figure 1). The most complex part of It is also important to note the three-dimensional (3D)
configurations of bivalents at metaphase I. This means that
sister chromatids (bound together by cohesin proteins) are
ELS subject area: Cell Biology all strictly positioned in the equatorial plate, facilitating
cleavage and transportation of the individual (recombined)
How to cite: chromosomes to opposite poles, once sister chromatid
Hultén, Maj A (September 2010) Meiosis. In: Encyclopedia of Life
cohesion along chromosome arms lapses at the onset of
Sciences (ELS). John Wiley & Sons, Ltd: Chichester.
DOI: 10.1002/9780470015902.a0005772.pub2
anaphase I (Figure 1). This type of reductional segregation is
allowed because sister chromatid cohesion is actively

ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net 1
Meiosis

Table 1 Prophase I: stages and landmark events of chromosome behaviour


Stage Description
Leptotene Chromatin condensation begins and the chromosomes first become visible as long, extended thread-
like structures. As part of this condensation process, the chromosomes form a proteinaceous core
(axial element) composed of cohesion proteins and two synaptonemal complex proteins, SYCP2 and
SYCP3. The telomeres (the DNA–protein complexes that cap the ends of all chromosomes) bind to
the nuclear membrane and begin to localise within a limited area of the membrane (‘bouquet’
formation)
Zygotene The transition from leptotene to zygotene is marked by the start of chromosome pairing. The central
core of the synaptonemal complex is formed by the protein SYCP1. Synapsis generally initiates from
chromosome ends. DNA repair proteins, such as RAD51 and DMC1, decorate unpaired axes until
synapsis is complete (see Figure 4)
Pachytene By convention, zygotene ends and pachytene commences at the point when maximal synapsis is
achieved. Intermediate recombination structures mature into crossovers in this stage. The positions of
exchange are marked by foci of the mismatch repair protein MLH1 (see Figure 6)
Diplotene Transition to the diplotene stage is signalled by the lapsing of homologous chromosome pairing, a
process termed desynapsis. Human foetal oocytes arrest in diplotene and remain in this stage until
shortly before ovulation
Diakinesis Desynapsis is complete and the bivalents are held together only at the points of reciprocal
recombination/crossing-over, now visible as chiasmata. The chromosomes continue to condense

protected around the centromeric regions of sister chro- alignment, although in fungi, animals and plants, there
matids. The orientation of paternal and maternal homo- appears to exist both an early recombination-independent
logues at the metaphase I plate is random. Therefore, and DNA double-strand break (DSB)-dependent com-
although each cell produced by meiosis contains only one ponents (Zickler, 2006). It seems possible that in humans
of each homologue, the number of possible combinations this alignment is mediated by repetitive DNA sequences
of maternal and paternal homologues is 2n, where n is the such as the so-called minisatellites. Second, after alignment
haploid number of chromosomes. The random assortment of homologous chromosomes (or chromosomal segments),
of homologues at the metaphase I stage produces in the chromosomes associate intimately in a process termed
humans 223 (8 388 608) different combinations of chromo- synapsis (Figure 4; Barlow et al., 1997). Synapsis is mediated
somes. An additional important mechanism for genetic by a meiosis-specific proteinaceous structure, the synap-
diversity in offspring is based on the reciprocal recombin- tonemal complex (SC) (review in Page and Hawley, 2004).
ation that has taken place between their grandparental There is some evidence that the initial phase of synapsis at
chromosomes during meiosis in ovaries and testes of their early pachytene is limited to regions of chromosome
parents (Figure 1). Bearing in mind that the segregation of homology (homosynapsis), but this restriction may ease
sister chromatids at anaphase II is also random, each child later to allow some non-homologous synapsis (hetero-
is expected to be genetically unique. synapsis). This change in synaptic behaviour is most easily
identified in cases where there are heterozygosities for
chromosome rearrangements, such as in translocations
Chromosome Pairing of Homologous and inversions (Figure 3; Saadallah and Hultén, 1986).
Meiotic homologous chromosome pairing and crossover
Parental Chromosomes at Meiosis I formation in ovaries of mammals take place during foetal
development (Lyrakou et al., 2002; Perheentupa and
Homologous chromosomes are usually located in separate Huhtaniemi, 2009). In contrast, this is a lifelong process
regions of the premeiotic nucleus. Chromosomes, there- ongoing from puberty in the testes (Perheentupa and
fore, need to move actively around the nucleus to search for Huhtaniemi, 2009).
and identify their homologous partner. Meiotic chromo-
some pairing takes place at the leptotene and zygotene
stages and is by definition completed at the pachytene
stage. It is generally considered to be a two-phase process
Tools for Identification of
(Figure 2, Figure 3, Figure 4, Figure 5, Figure 6 and Figure 7). Meiosis-specific Chromosome Structures
First, homologous chromosomes (or chromosomal seg-
ments) become aligned with chromosome ends attached to Chromosome preparations made in the traditional way
the nuclear membrane. Currently, it is not clear how dis- (by hypotonic pretreatment and Carnoy fixation) from,
tance recognition of homology is achieved to enable this for example, biopsies of mice and human testis allow

2 ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net
Meiosis

Chromatin

LE CE
Kinetochore

(a)

Metaphase I Anaphase I Chromatin


Cohesin
axis
Transverse
fibrils Lateral
(central (axial)
element) element
(b)

(c)
Telophase II Metaphase II
Figure 2 (a) Electron microscopy (EM) image of a single synaptonemal
Figure 1 Chromosome behaviour in the later stages of meiosis. After complex (SC). The tripartite structure of the SC is evident. The structure
diakinesis, the bivalents attach to spindle microtubules and orient in a comprises two lateral elements (LEs) each of 50 nm and a central region of
monopolar fashion at metaphase I. When orientation is complete, the 100 nm. A thin central element (CE) can be seen within the central region.
cohesin complex proteins that bind sister chromatids are removed (except (b) Cartoon to show the relationship of the chromatin to the axial cores
at the kinetochores), releasing the chiasmata and allowing the formed by the cohesin complex proteins and the LEs of the SC. Two SC
chromosomes to be pulled to the spindle poles at anaphase I. The proteins (SYCP2 and SYCP3) are present in the LEs. These are large proteins
chromosomes reorient on the metaphase II spindle, and the chromatids that associate with the cohesin complex holding sister chromatids together.
separate and move to opposite spindle poles. The net result is the The drawing shows the cohesin constitution of the LE separately, but it is to
formation of cells containing a haploid, unreplicated genome. See also be noted that there is no indication that there are in fact two layers making
Figure 7a as regards the reciprocal recombination/crossing-over/chiasma up the LE. Only one protein, SYCP1, has so far been isolated from the
formation taking place at prophase I of meiosis. central region. The C-terminal domain of this protein has been shown to
bind to the chromosomal axes. The central part of the protein is a coiled
coil and stretches out into the central region of the SC. The protein does
identification of numerous sperm-producing cells (sperm- not traverse the whole central region but overlaps and associates, at the
atocytes) at the meiotic pairing stage of prophase I, as well N-terminal domain, with SYCP1 fibrils stretching out from the opposite
as much more rarely cells at the metaphase I and metaphase lateral element. This overlapped region appears to be largely responsible for
II stages. Interestingly, human prophase chromosomes at the central element. (c) EM of an SC stained to show recombination
nodules (arrows). (a) and (c) courtesy of N. Sadaallah.
the fully synapsed stage (pachytene) show ‘spontaneous’
banding patterns (so-called chromomeres) following block
staining by Giemsa, for example. This banding pattern technology alone and only a few groups have used both EM
corresponds perfectly to the G-banding on mitotic and IF for the analysis of SCs on the same material of testes
chromosomes as seen in ordinary lymphocyte preparations or ovaries (Barlow and Hultén, 1997); and there is so far no
following trypsin treatment and used in clinical cyto- report applying all three approaches including prepar-
genetics service laboratories. In 1956, it was discovered that ations made in the traditional way by hypotonic pretreat-
following surface spreading, electron microscopy (EM) ment and Carnoy fixation. See also: Chromosomal Bands
analysis would allow the detailed visualisation of the dif- and Sequence Features; Karyotype Analysis and Chromo-
ferent components of this pairing structure, termed the some Banding
synaptonemal complex (Moses, 1956). Subsequently, it
was found that immunofluorescence (IF) analysis, apply- EM analysis of SCs
ing various antibodies against proteins identified as com-
ponents of the SC, provided much more detailed The SC was first identified by EM analysis of surface-
information on the organisation of the SC than that spread pachytene spermatocytes, using silver or phospho-
obtained by EM analysis alone (examples in Figure 4 and tungstic acid (PTA) staining (Moses, 1956). It appears as a
Figure 6). Most researchers have since applied the IF tripartite ribbon of proteins running along the axes of

ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net 3
Meiosis

(a) (b)

Figure 3 Electron microscopy (EM) pictures of synaptonemal complexes (SCs) at the pachytene stage showing so-called excrescences of unpaired
chromosome segments. (a) Unpaired chromosome segments within inverted segment of chromosome 13 show excrescences. The nucleolar organising
region (NOR) has been broken into two separate parts (N) in the formation of the inversion. (b) Interlocked SC where the unpaired segments show the same
excrescences as the unpaired segments of the XY bivalent, marked X and Y. Only a very small part of the short arms of the X and Y are synapsed. Courtesy of
N. Sadaallah.

Figure 4 Immunofluorescence microscopy (IM) picture of human spermatocyte at the zygotene stage illustrating the RAD51 foci. (a) Labelling with
anti-hRAD51 (white) and sera GS (centromeres, red) show numerous RAD51 foci, some indicated by arrows. (b) Same picture as in (a) but with anti-A1
(anti-lateral element, white) superimposed, illustrating how the RAD51 foci have disappeared along the segments of two bivalents, where synapsis has been
successful, but remain within the interstitial unpaired segment (green and white arrows in (a) and (b) and white arrow in insert). Reproduced from Barlow
et al. (1997), with permission from Nature Publishing Group.

paired chromosomes (Figure 2). Any unpaired segments and the disrupted meiotic cDNA 1 (DMC1) proteins. SC
show so-called excrescences (Figure 3; Saadallah and lengths at pachytene vary considerably between human
Hultén, 1986), indicative of aggregation of proteins that sperm-producing (spermatocyte) and egg-producing
would disappear following synapsis, including the histone (oocyte) cells: in spermatocytes, the total length per cell is
2 variant AX (gH2AX), the radiation sensitive 51 (RAD51) approximately 260 mm, whereas in oocytes, it exceeds

4 ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net
Meiosis

5 3
3 5
3 5
5 3
SPO11
Double-strand break

MRE11/RAD50/NBS1
Strand resection

RPA, RAD51, DMC-1

Single end invasion

Second end capture

MSH4/MSH5

Double Holliday
junction

MLH3/MLH1, GEN1

Crossover
formation

Figure 5 Double-strand break (DSB) model of recombination (see Kirkpatrick, 1999). Cutting of strands in opposite orientation at each junction
(arrowhead) produces a crossover.

520 mm (Wallace and Hultén, 1985; Tease and Hultén, Electron microscope investigations also identified small
2004). In both sexes, a limited part of each chromosome is structures, termed recombination nodules, associated with
associated with the SC and, therefore, in intimate contact SC formation. These nodules are categorised as early
with the homologous regions on its partner chromosome. recombination nodules (ENs), transformed nodules (TNs)
Most of the chromosome is instead present as chromatin or late recombination nodules (LNs) (Moens et al., 2007).
loops surrounding the SC and is not involved in the This discrimination is based on the time of appearance of
complex interactions between two parental chromatids, the recombination nodules and presumptions regarding
leading up to reciprocal recombination/crossing-over/chi- function. ENs are associated with synapsing chromosomal
asma formation. However, recombination is thought to be axes. They are generally small and ellipsoid and are
initiated by the formation of DSBs in the DNA loops. Then assumed to be involved in homology testing between
these breaks are brought to the chromosome axis where aligned chromosomal segments. TNs are formed from ENs
they are further processed to allow meiotic recombination. through the acquisition of various DNA repair proteins.
As far as is known, there are no specific DNA sequences These nodules are associated with SC extension as part of
localised within the central region of the SC, but this DNA the progression of synapsis. LNs are present following
tends to be more AT (adenosine, thymidine) rich than the synapsis (Figure 2c; Saadallah and Hultén, 1986). They can
genome at large. vary in appearance from round structures to bar-like

ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net 5
Meiosis

objects straddling the SC and are believed to be involved


in the final part of the sequence of events leading up to
reciprocal recombination. ENs occur in larger numbers
than LNs, typically at least an order of magnitude higher. It
is generally held that LNs evolve from a subset of ENs.

Immunofluorescence analysis of the


structural components of the SC
The first meiotic proteins visualised are the components of
the cohesin complex, which holds sister chromatids toge-
ther, that is the structural maintenance of chromosome 1b
(SMC1b), the recombination 8 (REC8) and the stromaline
3 (STAG3) proteins (Prieto et al., 2004). The proteins
(a) appear as punctuated foci in preleptotene cells. During
leptotene, these signals organise into fibrillar patterns and
proteins that make up the SC appear. The cohesins attach
to the chromatin and become sandwiched within the axial
elements and the proteins forming the remaining part of the
SC scaffold (Figure 2). Three major SC proteins that form
the basis for the SC structure have been identified to date
(Table 2). Two of these, SYCP2 (synaptonemal complex
protein 2) and SYCP3 are located at the axes of chromo-
somes as they condense during early prophase I, forming
the axial/lateral elements of the SC. SYCP2 and SYCP3
appear to associate with (or are loaded on to) the cohesin
protein complex that binds together sister chromatids fol-
lowing DNA replication (Figure 2b). The third protein,
SYCP1, is present in the central region of the SC. SYCP1 is
a long filamentous protein. Its C-terminal end binds to the
(b) lateral element, with the N-terminal region pointing into
the central region. N-terminal regions from opposite lateral
elements associate centrally to hold the SC together (Figure
2b). More recently, other components of the central elem-
ent have been identified in mammals: SC central element
(SYCE) 1 and 2 plus FKBP6 (FK506 binding protein 6).

Immunofluorescence analysis of XY
behaviour at prophase I
IF analysis has allowed detailed information to be obtained
on the behaviour of aspects of specialised meiotic
chromosome pairing. For example, in human spermato-
cytes, the heteromorphic sex chromosomes undergo a
different schedule of synapsis from the autosomes (Arm-
strong et al., 1994). This pattern of behaviour may be
(c) predicated on the need to maintain the X in a genetically
inactive state during meiosis. The X- and Y-chromosomes
Figure 6 Detecting the positions of crossovers at pachytene using the
share a relatively small region of homology (approximately
DNA mismatch repair protein, MLH1. The cells have been stained using 2.6 Mb in length) at the distal ends of their short arms (the
antibodies against SYCP3 (red), MLH1 (yellow) and, in the spermatocyte, ‘pseudoautosomal’ region, PAR). They also have a much
the kinetochore (blue). (a) Normal pachytene spermatocyte. (b) Normal smaller (approximately 0.23 Mb) PAR of homology at the
pachytene oocyte. There are obvious differences between the spermatocyte
and the oocyte: the SCs are much longer in the oocyte; there are more
ends of the long arms. Synapsis initiates at the short-arm
MLH1 foci in the oocyte; MLH1 foci tend to be positioned closer to the PAR in early pachytene and goes beyond the region of
telomeres in the spermatocyte. (c) Pachytene oocyte showing failure of homology occasionally to include the entire short arm.
pairing affecting some chromosomes (arrows). MLH1 foci are present on However, this heterosynapsis is not long lasting and the
synapsed chromosome pairs (arrowheads). chromosomes begin to separate (desynapse). By mid-
pachytene, the chromosomes are associated only at their

6 ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net
Meiosis

x x 1

3
XY
(a) (b)

(c) (d)

Figure 7 (a) Cartoon illustrating how the configurations in (b) are achieved by bivalents with one or two chiasmata. Two chromosome pairs are shown, in
each case one homologue is coloured blue, the other red. A reciprocal recombination/crossover between a blue and a red chromatid generates an exchange
that is visible as a cross-configuration/chiasma in the metaphase I bivalent. The acrocentric chromosome pair has a single reciprocal recombination/
crossover/chiasma and produces a rod bivalent at metaphase I; the metacentric has two reciprocal recombinations/crossovers/chiasmata and produces a ring
bivalent. (b) Metaphase I spermatocyte. Pairs of homologous chromosomes are held together by variable numbers of chiasmata. Bivalents with 1, 2 or 3
chiasmata are indicated, as is the XY-chromosome pair. (c) Metaphase II spermatocyte. The chromatids of each chromosome (arrowheads) are held together
only at the centromeres (arrow). (d) Metaphase I spermatocyte from a man with an abnormally low number of chiasmata. In contrast to (b), bivalents
(arrows) have only one or two chiasmata; additionally, a number of chromosome pairs have failed to form crossovers and are present as univalents
(arrowheads).

Table 2 Major components of the mammalian synaptonemal complex and cohesin complex
Axial/lateral element Transverse filament Central element Cohesin core
SYCP2 SYCP1 TEX12 STAG3
SYCP3 SYCE1 SMC1b
SYCE2 REC8

very distal tips of their short arms. In approximately half of An Integrated Molecular and
early pachytene cells, the regions of homology of the long
arms also associate. The sex chromosomes undergo con-
Cytogenetic Model of the
siderable morphological changes during pachytene, even- Crossing-over Process
tually forming a highly differentiated condensed ‘sex
vesicle’ in late pachytene. This unusual pattern of synapsis The currently accepted molecular model of crossing-over
and chromosome differentiation provides a reliable means is the DSB repair (DSBR) model (Kirkpatrick, 1999).The
to subdivide pachytene in the human male, and thereby can DSBR pathway relies on double Holliday junction (dHJ)
be exploited to distinguish early to late pachytene cells formation and resolution to produce crossovers (Figure 5).
when examining meiotic behaviour of autosomes. The This includes DNA breaks initiated at an early stage of
X-chromosome pair in human prophase I oocytes does not meiosis (leptotene) followed by reciprocal recombination
show any difference in behaviour from the autosomes. between parental chromatids, which is completed during

ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net 7
Meiosis

Table 3 Timing and possible functions of some DNA repair proteins that associate with chromosome cores during leptotene,
zygotene and pachytene stages of prophase I
Protein Stages present Role
SPO11 (sporulation) Leptotene to zygotene Produces DSBs, essential for synapsis
RAD51 Leptotene to pachytene RecA homologue that binds to
single-stranded DNA; possible role in
early steps of recombination by
mediating single end invasion
DMC1 (disrupted meiotic cDNA) Leptotene to pachytene RecA homologue that binds to
single-stranded DNA; colocalises
with Rad51
RPA (replication protein A) Zygotene to pachytene Binds to single-stranded DNA,
possibly involved in early steps of
recombination
BLM (Bloom syndrome mutated Leptotene to pachytene Helicase, colocalises with RPA,
helicase) possibly functions in recombination
MSH 4/5 (MutS homologue) Zygotene to pachytene Required for chromosome pairing; in
yeast, interacts with MLH1 to control
crossing-over
MLH1 (MutL homologue) Zygotene to diplotene Required for crossing-over
MLH3 Pachytene Involved in DNA mismatch repair
GEN1 Pachytene Required for resolving of Holliday
junctions

the following zygotene and pachytene stages (Table 1). This thus likely constituents of the excrescences of unpaired
model is primarily based on experiments in bacteria and lateral elements of the SC seen by EM (Figure 3; Saadallah
yeast, but is likely to be equally relevant to higher organ- and Hultén, 1986). The majority of these DSBs will not go
isms, including humans (Sung and Klein, 2006; West, on to become involved in crossing-over; instead, the
2009). An expanding array of DNA repair proteins have invading strand is displaced and reanneals with the other
been identified as playing a role in the recombination side of the DSB. This is known as synthesis-dependent
process of crossing-over (Table 3). strand annealing and can produce gene conversion events,
involving transition of DNA nucleotides between maternal
and paternal chromosomes. It is likely that the numerous
SPO11 induces double-strand breaks at the DSBs constitute sites of potential crossovers reflected as
leptotene stage the type of hotspots described by the model proposed by
sperm experimentation. This model implies that there are
DSBs appear to be an essential prerequisite to homology so-called crossover hotspots of around 1–2 kb with inter-
testing between parental chromosomes, which involves the vals of lower recombination frequency within the adjacent
initial stages of recombination between closely aligned but 90–125 kb (review in Kauppi et al., 2009; Hastings et al.,
not yet synapsed chromosomes. Synapsis thus requires the 2009; Stankiewics and Lupski, 2010). It is important to
formation of DSBs in the chromatin through the action of note, however, that only a small proportion of these will be
the topoisomerase II-related protein SPO11 (sporulation selected for crossover formation, as described in the fol-
gene 11) during leptotene. An endonuclease then nicks the lowing text.
DNA close to the DSB liberating SPO11 attached to a short
oligo nucleotide (Keeney and Neale, 2006). By this action,
the first bases of single-stranded DNA (ssDNA) overhangs MRE11 is required for recombination
are formed. They are extended by a combination of activ- initiation
ities involving exonucleases and helicases.
Numerous DSBs are formed at leptotene and it is widely In addition to SPO11, a number of other proteins are
accepted that the appearance of g-H2AX, a variant histone required for DSB formation and recombination initiation.
phosphorylated at serine 139, is a suitable marker for their The best studied of these accessory proteins are the MRX
presence (Redon et al., 2002). Foci of g-H2AX can be and MRN complexes. The MRN complex, composed of
detected by immuno-cytohistochemistry. These foci dis- the meiotic recombination 11 (MRE11), the RAD50 and
appear following synapsis but remain visible along the the Nijmegem breakage syndrome 1 (NBS1) proteins
length of any unpaired lateral element (including the localises to sites of DNA damage where it plays a role in
unpaired parts of the XY bivalent). The g-H2AX foci are genome maintenance (Rupnik et al., 2010). MRE11 has

8 ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net
Meiosis

multiple functions including 3’ to 5’ double-strand (ds) and recombination as part of the ongoing crossover and chiasma
single-strand (ss) DNA exonuclease and ssDNA endonu- formation process. A subset (approximately 50 in human
clease activities (Borde, 2007); RAD50 contains Zn2+- males, 70 in human females) is resolved as crossovers while
binding hooks that allow dimerisation, and the formation the remainder is resolved as gene conversion events, thus
of a flexible bridge that can cross a DSB gap or link sister contributing to the genetic diversity in offspring. See also:
chromatids together. Combined with MRE11 and NBS1 Genetic Mapping: Comparison of Direct and Indirect
dimers, the complex can be tethered to DNA. Approaches; Genetic Maps: Direct Meiotic Analysis
During meiosis, the MRX complex is required for
SPO11-mediated DSB formation. The MRN complex is
also believed to be involved in recombination initiation, Digestion and unwinding DNA by a number
specifically 5’ to 3’ exonucleolytic resection. The exact of additional proteins
mechanism for this is unknown but it has been suggested
Following on from the formation of a displacement loop,
that the MRN complex recruits and controls other exo-
the joined DNA molecules must be arranged and held in
nuclease activity. These exonucleases degrade the ends of
the correct conformation for dHJ formation. It is believed
the DSB to produce ssDNA overhangs.
that the components of the TNs, identified by EM analysis
of the SC, are responsible for this although the exact
RAD51 and DMC1 promote DNA strand mechanism of action is unknown. These proteins include
invasion endonucleases for digesting DNA and also helicases such
as BLM (Bloom Syndrome) for unwinding DNA. BLM is
Following strand resection, the ssDNA overhangs are then
of particular importance as the C-terminal domain can
available for binding by RAD51 and DMC1, which pro-
interact with a number of proteins, including MLH1 (mutL
mote strand invasion of a chromatid from the homologous
homologue 1), which may represent a method for recom-
chromosome (Figure 4; Barlow et al., 1997; Keeney and
bination nodule progression towards finalising the cross-
Neale, 2006). Stable, homologous strand invasion dis-
over process (see later discussion).
places the homologous strand to form a displacement loop,
Other components of the TNs have been confirmed as
which is subsequently available for capture by the invading
RPA (replication protein A), MSH4 (mutS homologue 4)
homologue thus forming a mature dHJ. The RAD51/
plus various topoisomerases. RPA is a heterotrimeric
DMC1 recombinases thus promote reciprocal strand
protein complex, which can bind to ssDNA. RPA associ-
exchange between parental chromatids and lead to intim-
ates with chromosome cores of synapsing homologues and
ate pairing and synapsis of the lateral elements of the SC.
overlaps with RAD51 in timing of appearance. BLM is a
This zipping-up process of homologous chromosomes at
DNA helicase that is capable of unzipping dsDNA. All
the zygotene stage most often starts near the ends of
eukaryotes studied so far also encode homologues of the
chromosomes and progresses towards the middle. In some
baterial mismatch repair (MMR) proteins, MutL and
organisms, recombination sites fated to become crossovers
MutS (methyl-directed mismatch repair proteins). The two
are thought to act as sites of synaptonemal complex (SC)
meiosis-specific MutS homologues MSH4 and MSH5
initiation (Lynn et al., 2007). When full synapsis is
function together as a heterodimer to facilitate the for-
achieved, the pachytene stage has been reached.
mation of reciprocal recombination/crossing-over/chi-
Studies have indicated that RAD51 and DMC1 are
asmata formation. The MSH4–MSH5 dimer is not
components of ENs as identified by EM analysis (Moens
involved in MMR but is believed to function as a sliding
et al., 2007). RAD51 and DMC1 are homologues of the
clamp capable of stabilising Holliday junctions.
bacterial RecA (recombinational DNA repair gene A)
protein, which is involved in homologous recombination
and repair of DNA damage. IF analysis has illustrated how Finalising the crossing-over process by the
RAD51 is initially deposited as discrete foci on one or other action of MLH3/MLH1 and GEN1
of the unsynapsed lateral elements of the SC. These foci
disappear once synapsis is established but remain in any There are also several MutL homologues in eukaryotes.
unsynapsed regions (including the unsynapsed regions of The DNA MMR protein MLH1 and the meiosis-specific
XY) and are likely to represent parts of the SC excrescences MLH3 have been shown to be present in the LNs as rec-
seen by EM (Figure 3; Saadallah and Hultén, 1986). ognised by EM, marking foci of crossovers (Figure 2; see
Apparently, the appearance and disappearance of RAD51 also Saadallah and Hultén, 1986). Therefore, the selected
is a rapid process as the RAD51 foci can be seen to have sites of crossover formation can be identified at pachytene
come and gone within the same synapsing bivalent during by IF staining for the presence of the DNA MMR proteins,
zygotene (Figure 4; Barlow et al., 1997). MLH1 and MLH3. The presence of MLH1 as discrete foci
In most organisms studied in this respect, the number of along the SC in human spermatocytes and oocytes is
DNA DSBs is much greater than the observed number of illustrated in Figure 6. These foci mimic the numbers and
crossovers. Thus, many initial recombination events are distributions of reciprocal recombination sites and chi-
not resolved as a crossover. Equally, most of the RAD51/ asmata (see later discussion). Recently, another protein,
DMC1-containing ENs are not involved in reciprocal GEN1, functioning in resolving the Holliday junctions has

ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net 9
Meiosis

XY

(a) (b)

Figure 8 Human spermatocyte at the metaphase I stages (a) and (b), where the chiasmata have been highlighted (middle) and oocyte at the metaphase I
stage, revised from Yuncken (1968). Note the difficulty in identifying the chiasmata in the oocyte in comparison to the spermatocyte. (a) Reproduced from
Hultén et al. (1970), with permission and (b) from Yuncken (1968), with permission from S. Kargel AG, Basel.

been discovered (review in West, 2009). See also: Mismatch crossovers along chromosome arms reflect at least two fac-
Repair Genes tors: preferential formation in certain chromosomal seg-
ments and positive crossover interference, a process by which
Chiasma(ta) the presence of a crossover reduces the probability of a sec-
ond crossover being formed nearby. The former is exempli-
The position of crossover events becomes cytogenetically fied by the high rate of chiasma formation close to
apparent as a bivalent condenses during diplotene and chromosome ends in human spermatocytes (Figure 6a and
diakinesis (Figure 7a and Figure 8). Sister chromatids remain Figure 7b). The influence of interference is most easily seen
tightly associated (due to the sister chromatid cohesion when two or more chiasmata are present along a chromo-
complex) proximally and distally to the crossover giving some arm. Adjacent chiasmata rarely form close to one
rise to a cytogenetically visible structure called chiasma another but are more usually separated by a comparatively
(plural chiasmata). Chiasma numbers and positions can be large chromosome length. Currently, the mechanism of
analysed in metaphase I spermatocytes (Figure 7b). Chi- interference is unclear (review in Stahl and Housworth,
asmata provide a reliable means of determining the rate of 2009; Martinez-Perez and Colaiacovo, 2009).
crossing-over at both the genomic and the chromosomal A third factor that affects the number and position of
levels in the human male (Laurie and Hultén, 1985; Hultén, crossovers is the presence of mechanisms that favour the
1994). See also: Genetic Maps: Direct Meiotic Analysis repair of DNA DSBs as non-crossover events. For
Unfortunately, this type of analysis has so far not been example, mice and yeast mutants lacking the BLM helicase
possible in human females (Figure 8). Female meiosis arrests display an increased number of chiasmata between hom-
at the diplotene stage in foetal ovaries, at which time ologous chromosomes (Holloway et al., 2010).
chromosomes are long and slender, and it is exceedingly
difficult to differentiate twists from chiasmata at this stage.
Metaphase I takes place just before ovulation, usually Sex differences in frequency and position of
within a single oocyte at a time. This scarcity of material for crossing-over/chiasmata
study has precluded the direct approach for studying pat-
terns of meiotic reciprocal recombination and crossing- Human males with normal fertility show remarkably little
over in human females by analysis of chiasmata at the interindividual variation in numbers of chiasmata, with an
metaphase I stage. However, these types of studies have average of approximately 50 per spermatocyte (range 43–60
been performed in mice, using hormone stimulation and per cell) (Hultén, 1974; Laurie and Hultén, 1985). Analysis of
in vitro culture of oocytes (Hultén et al., 1995; Lawrie et al., MLH1 foci at pachytene has produced a similar estimate of
1995). See also: Genetic Mapping: Comparison of Direct crossover frequency in spermatocytes (Barlow and Hultén,
and Indirect Approaches; Genetic Maps: Direct Meiotic 1998; Lynn et al., 2002; Sun et al., 2004; Martin, 2008).
Analysis Numbers of crossovers, as estimated from MLH1 foci, is
Crossovers are non-randomly distributed with regard to higher in oocytes prepared from foetal ovaries. Initial MLH1
both numbers per chromosome pair and positions along analysis has suggested an average of approximately 70
chromosome arms. Each bivalent receives at least one crossovers per oocyte at pachytene, with a larger intercell
exchange (the so-called obligate crossover). Additional variability (range 40–100) than in spermatocytes (Barlow
crossovers (up to four or five per bivalent) are more likely the and Hultén, 1998). Subsequent studies have confirmed the
longer the individual bivalents are at the stage when crossing- occurrence of a large variation in crossover frequency
over is finalised, that is, the pachytene stage. The positions of between individual oocytes (Tease et al., 2002; Lenzi et al.,

10 ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net
Meiosis

2005; Cheng et al., 2009). It is likely that some of this variation family of proteins until anaphase II, at which time REC8 is
is due to underestimation of the crossover frequency when cleaved by separase. The presence and retention of cohesin
counting MLH1 foci due to the temporary nature of their proteins is crucial for monopolar orientation of sister
appearance. The higher rate of recombination in oocytes is kinetochores (both attached to the same pole) at metaphase
most probably related to the considerable difference in I and the remaining centromeric cohesion allows the
pachytene chromosome length described earlier (Tease et al., chromatids to align at metaphase II.
2006). The female genome has a longer physical platform for A programmed lapse of sister chromatid cohesin along
establishment of reciprocal recombination/crossing-over/ chromosomal arms results in the loss of the crossover
chiasmata than the male. Not only do the two sexes show a associations holding the homologous chromosomes toge-
significant variation in recombination frequency, but they ther. The homologues are then able to move to opposite
also display differences in distribution (Tease et al., 2002). In spindle poles as the microtubules shorten. The resulting
spermatocytes, the MLH1 foci/chiasmata are often located daughter nuclei, therefore, obtain half of the chromosome
very close to the ends of the chromosomal axes. In oocytes, complement (Figure 7c). It is important to note that
MLH1 foci are located more interstitially (away from maternally and paternally inherited homologues assort at
chromosome ends) and only very rarely positioned so near to random at anaphase I. The resulting daughter cells,
telomeric segments as in spermatocytes (compare Figure 6a therefore, contain a haploid chromosome set different from
and b). Indirect family linkage analyses, tracing DNA that contributed by maternal and paternal germ cells. It is
markers between generations, have similarly identified this also important to remember that the chromatids of each
intersex variation in numbers and distributions of cross- chromosome are now no longer identical because of
overs (Lynn et al., 2004). See also: Genetic Mapping: reciprocal recombination (see Figure 1). These two pro-
Comparison of Direct and Indirect Approaches; Genetic cesses of random assortment and meiotic recombination
Maps: Direct Meiotic Analysis contribute to genetic variation in offspring.
In oocytes, the cytoplasmic division at anaphase I, tak-
ing place before ovulation, is highly unequal, producing a
Chromosome Segregation large secondary oocyte and a residual, minute cell termed
the first polar body. The polar body ceases meiosis and
At metaphase I, the pairs of homologous chromosomes the chromosomes begin to degenerate. Oocytes arrest at
(bivalents) attach to spindle microtubules and move to the metaphase II and are ovulated at this stage. They are
spindle equator in preparation for nuclear division. At the stimulated to complete meiosis only after fertilisation. In
first meiotic division, sister kinetochores (microtubule- spermatocytes, the products of the first meiotic division are
attaching segments of the centromeres) act as a single unit equal in size and both go on to complete meiosis II.
and orient in a monopolar fashion (both attached to the same Chromosome condensation resumes very shortly after
pole of microtubules). This is due to the fact that sister anaphase I, and the chromosomes soon orient on the
chromatid cohesion is actively protected around centromeric metaphase II spindle. During the second meiotic division,
regions during meiosis I, which is quite distinct from mitosis the release of cohesion along all chromosomal regions
where the kinetochores of sister chromatids orient to allows the segregation of the sister chromatids. Although
opposite spindle poles. See also: Chromosomes during Cell this second meiotic division has much in common with a
Division; Kinetochore: Structure, Function and Evolution; mitotic division, the daughter nuclei that are produced are
Mitosis: Chromosome Segregation and Stability not identical – due to reciprocal recombination at prophase
The chiasmata in each bivalent act as anchors to hold the I and due to the random orientation and assortment of
pairs of homologues together during the orientation pro- bivalents at anaphase I.
cess within the equatorial plate of metaphase I cells. Evi- Once chromosome orientation is complete (and following
dence from experimental organisms shows that incorrect fertilisation in the case of oocytes), centromere cohesion lapses
orientation, or failure to attach to spindle microtubules, and the spindle microtubules pull each chromatid to the poles
can switch on a signalling pathway that results in cell div- at anaphase II. Meiosis is now complete. The resulting cells
ision arrest until the chromosomes successfully interact in male meiosis are termed spermatids. They undergo very
with the spindle (Nicklas, 1997). It is presumed that a considerable morphological changes in the process of forming
similar mechanism exists in human spermatocytes. There is spermatozoa (spermiogenesis). In female meiosis, cytoplasmic
some doubt as to whether it is as sensitive (or possibly even division at anaphase II is again very unequal producing a
present) in human oocytes. second small polar body. The egg nucleus decondenses to
The cohesin protein complex is present at the kine- form the female pronucleus of the zygote. See also: Mitosis:
tochore of meiotic chromosomes (Suja and Barbero, 2009). Chromosome Segregation and Stability
Three meiosis-specific variants of cohesin components
have been identified: STAG3, SMC1b and REC8 (Table 2).
Sister chromatid cohesion is released from chromosomes Consequences of Abnormal Meiosis
by the action of the separase enzyme, ESP1 (extra spindle
pole bodies 1), at the metaphase to anaphase I transition. Meiosis is a surprisingly error-prone process in humans
Centromeric cohesion is protected by the Shugoshin (SGO) and thereby has a significant adverse impact on human

ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net 11
Meiosis

reproduction (review in Hultén et al., 2010a). Meiotic meiotic errors such as a significantly reduced number of
errors can result in infertility and are also responsible for MLH1 foci at the pachytene stage of prophase I or chiasma
generating the genetically imbalanced gametes that pro- frequency at metaphase I (Hultén et al., 1970; review in
duce aneuploid conceptions. Errors of meiotic chromo- Martin, 2008).
some behaviour are the single largest contributor to
genetically determined congenital abnormalities and Non-disjunction
mental impairment affecting live-born children.
Chromosome abnormalities occur at a much higher level in
Chromosome pairing and recombination humans than in any other species. More than 50–60% of
errors clinically recognised pregnancies have some form of
chromosome abnormality (Vanneste et al., 2009; Hultén
All aspects of meiotic chromosome behaviour are error et al., 2010a). Monosomies and most trisomies are nor-
prone. For example, over 30% of human pachytene mally eliminated early in gestation because they are gen-
oocytes may show some form of chromosome pairing erally incompatible with normal development. Sex
anomaly (Tease et al., 2006). Commonly, homologous chromosome monosomy (45, X) is the only monosomic
chromosomes fail to pair, a phenomenon termed asynapsis condition that may proceed to term. The consequences of
(Figure 6c). In yeast, anomalies of chromosome pairing can trisomy are less severe for the sex chromosomes compared
trigger cell checkpoints resulting in arrest of cell develop- with the autosomes, and therefore, these are more common
ment and ultimately cell death. It seems likely a similar among live births.
phenomenon occurs in human germ cells. If this mech- Analyses of the chromosome complements of sperm-
anism is present, then any increase in the already high atozoa have shown that malsegregation (non-disjunction)
proportion of foetal oocytes with pairing errors could of chromosomes is normally uncommon (review in Martin,
result in depletion of the germ cell population and lead to 2008). Current estimates indicate a rate of approximately
the premature onset of menopause in women. Similarly, 2% aneuploid spermatozoa. Paternal non-disjunction only
meiotic pairing errors in spermatocytes could result in an accounts for approximately 10% of all autosomal trisomies
insufficiency of sperm production in men. A reduction in but certain autosomal trisomies (e.g. 2 and 8) have an
germ cell population would be far more detrimental in increased incidence of paternally derived cases. Paternal
human females because they produce just 300–400 mature errors contribute more frequently to sex chromosome
oocytes in a lifetime compared with 300–400 million sperm aneuploidy and account for approximately 10% of 47,
produced daily in human males. XXX females; 50% of 47, XXY; all 47, XYY males and
Incorrect alignment (unequal synapsis) of homologues 80% of 45, X females. Recent studies have indicated that
in conjunction with reciprocal recombination/crossing- infertile men attending reproductive clinics for collection of
over may generate a large variety of deletions and dupli- sperm for intracytoplasmic sperm injection (ICSI) tend to
cations that can cause genetic disease in offspring. It should have increased rates of aneuploidy in their sperm.
be noted, however, that some of this type of non-homolo- The rate of aneuploid sperm is elevated in men carrying
gous recombination (NAHR) is also thought to contribute structural chromosome rearrangements. For example, in
to the large amount of DNA copy number variation (CNV) men with reciprocal translocations, the proportion of gen-
that is now known to exist in the normal population (review etically imbalanced sperm may reach 60–70%. Homolo-
in Hastings et al., 2009; Sharp, 2009; Stankiewics and gous chromosome synapsis involving all four chromatids
Lupski, 2010). leads to a quadrivalent at metaphase I. The proportion of
Failure of crossing-over in fully synapsed chromosome chromosomally unbalanced sperm generated is dependent
pairs results in the presence of unpaired chromosomes on the way the quadrivalent segregates at anaphase I.
(univalents) at metaphase I (Figure 7d). This phenomenon is Commonly a chiasma is in an unusual position, between the
termed desynapsis. The lack of a chiasmate association centromere and the break point (the interstitial segment).
means that homologous chromosomes may segregate at Even the most normal (alternate) segregation pattern (where
random with respect to one another, with a resulting the normal and translocated chromosomes segregate to
marked increase in the rate of aneuploid gametes, which are opposite poles) will then give rise to unbalanced sperm. It
imbalanced as regards chromosome copy numbers. Any should in this context be noted that this complication means
unpaired chromosomes at metaphase I may undergo a so- that it is not possible from the proportion of sperm with
called predivision, that is, chromatids separate (as in normal, balanced and unbalanced chromosomes to differ-
mitosis) leading to extra and missing chromatids in entiate between alternate anaphase I segregation with a
daughter cells (see Hultén et al., 2008, 2010b). chiasma in the interstitial segment and adjacent segregation
In spermatocytes, there is evidence that failure of without a chiasma in this particular position (Goldman and
crossing-over can result in the developmental arrest of the Hultén, 1993; Armstrong and Hultén, 1998).
cells at early prophase and at metaphase I; these cells may Human oocytes have a higher predisposition for
then be lost through either necrotic or apoptotic pathways. chromosome segregation errors than spermatocytes. Over
Investigation of testicular biopsies from men attending 90% of conceptions with autosomal aneuploidy such as
infertility clinics have shown an increased likelihood of trisomy 21 associated with Down syndrome are the result

12 ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net
Meiosis

of chromosome non-disjunction in oocytes. Indirect gen- using fluorescence in situ hybridisation. Cytogenetics and Cell
etic analysis has indicated that abnormal patterns of Genetics 63: 16–23.
crossover distribution, that is, failure of crossing-over or Hastings PJ, Lupski JR, Rosenberg SM and Ira G (2009) Mech-
formation of exchanges very close to the centromere or anisms of change in gene copy number. Nature Reviews. Gen-
telomere, may increase the risk of chromosome non-dis- etics 10: 551–564.
junction in oocytes. It is not yet clear why, for some Holloway JK, Morelli MA, Borst PL and Cohen PE (2010)
chromosome pairs, bivalents with particular chiasma dis- Mammalian BLM helicase is critical for integrating multiple
tributions should be at increased risk of malsegregation. pathways of meiotic recombination. Journal of Cell Biology
The only factor that is known unequivocally to influence 188(6): 779–789.
Hultén M (1974) Chiasma distribution at diakinesis in the normal
the rate of non-disjunction is maternal age. Although many
human male. Hereditas 76(1): 55–78.
hypotheses have been advanced to explain the increased
Hultén M (1994) Chiasma formation, crossing-over and recom-
risk on chromosome segregation errors with age, it is fair to
bination in meiosis. Trends in Genetics 10(4): 112–113.
say that our understanding of this phenomenon remains Hultén M, Eliasson R and Tillinger KG (1970) Low chiasma
very incomplete. One possibility is the accumulation of pre- count and other meiotic irregularities in two infertile 46,
existing trisomy 21 oocytes during development from XY men with spermatogenic arrest. Hereditas 65(2):
foetal life to adulthood (Hultén et al., 2008, 2010b). We 285–290.
have documented that trisomy 21 mosaicism is common in Hultén MA, Patel S, Jonasson J and Iwarsson E (2010b) On the
foetal ovaries, obtained from termination of pregnancy for origin of the maternal age effect in trisomy 21 Down syndrome:
a non-medical/social reason (Hultén et al., 2008). We have the Oocyte Mosaicism Selection (OMS) model. Reproduction
further suggested that contrary to current dogma, the 139: 1–9.
maternal age effect is due to obligatory, secondary, non- Hultén MA, Patel SD, Tankimanova M et al. (2008) On the origin
disjunction of trisomy 21 oocytes that have accumulated of trisomy 21 Down syndrome. Molecular Cytogenetics 1: 21.
during development, involving a reduction in numbers Hultén MA, Smith E and Delhanty JDA (2010a) Errors in
from a prenatal maximum of 7 million to the 300–400 chromosome segregation during oogenesis and early embryo-
selected for maturation to ovulation in adulthood (Hultén genesis. In: Carrell DT and Peterson CM (eds) Reproductive
et al., 2010b). See also: Meiosis and Mitosis: Molecular Endocrinology and Infertility: Integrating Modern Clinical and
Control of Chromosome Separation Laboratory Practice. New York: Springer.
Hultén MA, Tease C and Lawrie NM (1995) Chiasma-based
genetic map of the mouse X chromosome. Chromosoma 104:
223–227.
References Kauppi L, May CA and Jeffreys AJ (2009) Analysis of meiotic
recombination products from human sperm. Methods in
Armstrong SJ and Hultén MA (1998) Meiotic segregation Molecular Biology 557: 323–355.
analysis by FISH investigations in sperm and spermatocytes of Keeney S and Neale MJ (2006) Initiation of meiotic recombin-
translocation heterozygotes. European Journal of Human Gen- ation by formation of DNA double-strand breaks: mechanism
etics 6: 430–431. and regulation. Biochemical Society Transactions 34(part 4):
Armstrong SJ, Kirkham AJ and Hultén MA (1994) XY 523–525.
chromosome behaviour in the germ-line of the human male: a Kirkpatrick DT (1999) Roles of the DNA mismatch repair and
FISH analysis of spatial orientation, chromatin condensation nucleotide excision repair proteins during meiosis. Cellular and
and pairing. Chromosome Research 2(6): 445–452. Molecular Life Sciences 55(3): 437–449.
Barlow AL, Benson FE, West SC and Hultén MA (1997) Distri- Laurie DA and Hultén MA (1985) Further studies on bivalent
bution of the Rad51 recombinase in human and mouse chiasma frequency in human males with normal karyotypes.
spermatocytes. EMBO Journal 16(17): 5207–5215. Annals of Human Genetics 49(part 3): 189–201.
Barlow AL and Hultén MA (1997) Sequential immunocytoge- Lawrie NM, Tease C and Hultén MA (1995) Chiasma frequency,
netics, molecular cytogenetics and transmission electron distribution and interference maps of mouse autosomes.
microscopy of microspread meiosis I oocytes from a human Chromosoma 104(4): 308–314.
fetal carrier of an unbalanced translocation. Chromosoma Lenzi ML, Smith J, Snowden T et al. (2005) Extreme hetero-
106(5): 293–303. geneity in the molecular events leading to the establishment of
Barlow AL and Hultén MA (1998) Crossing over analysis at chiasmata during meiosis I in human oocytes. American Journal
pachytene in man. European Journal of Human Genetics 6(4): of Human Genetics 76(1): 112–127.
350–358. Lynn A, Ashley T and Hassold T (2004) Variation in human
Borde V (2007) The multiple roles of the Mre11 complex for meiotic recombination. Annual Review of Genomics and Human
meiotic recombination. Chromosome Research 15: 551–563. Genetics 5: 317–349.
Cheng EY, Hunt PA, Naluai-Cecchini TA et al. (2009) Meiotic Lynn A, Koehler KE, Judis L et al. (2002) Covariation of
recombination in human oocytes. PLoS Genetics 5(9): e1000661 synaptonemal complex length and mammalian meiotic
Epub September 18. exchange rates. Science 296(5576): 2222–2225.
Goldman ASH and Hultén MA (1993) Analysis of chiasma Lynn A, Soucek R and Börner GV (2007) ZMM proteins during
frequency and first meiotic segregation in a human male meiosis: crossover artists at work. Chromosome Research 15:
reciprocal translocation heterozygote, t(1;11) (p36.3;q13.1), 591–605.

ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net 13
Meiosis

Lyrakou S, Hultén MA and Hartshorne GM (2002) Growth Tease C, Hartshorne GM and Hultén MA (2002) Patterns of
factors promote meiosis in mouse fetal ovaries in vitro. meiotic recombination in human fetal oocytes. American
Molecular Human Reproduction 8(10): 906–911. Journal of Human Genetics 70(6): 1469–1479.
Martin RH (2008) Cytogenetic determinants of male fertility. Tease C and Hultén MA (2004) Inter-sex variation in synapto-
Human Reproduction Update 14(4): 379–390. nemal complex lengths largely determine the different recom-
Martinez-Perez E and Colaiacovo MP (2009) Distribution of bination rates in male and female germ cells. Cytogenetic and
meiotic recombination events: talking to you neighbors. Genome Research 107(3–4): 208–215.
Current Opinion in Genetics & Development 19(2): 105–112. Vanneste E, Voet T, Caignec C et al. (2009) Chromosome
Moens PB, Marcon E, Shore JS, Kochakpour N and Spyropoulos instability is common in human cleavage-stage embryos. Nature
B (2007) Initiation and resolution of interhomolog connections: Medicine 15(5): 577–583.
crossover and non-crossover sites along mouse synaptonemal Wallace BM and Hultén MA (1985) Meiotic chromosome pairing
complexes. Journal of Cell Science 120(part 6): 1017–1027. in the normal human female. Annals of Human Genetics 49(3):
Moses MJ (1956) Chromosome structures in crayfish spermato- 215–226.
cytes. Journal of Biophysical and Biochemical Cytology 2: West SC (2009) The search for a human Holliday junction resol-
215–218. vase. Biochemical Society Transactions 37: 519–526.
Nicklas RB (1997) How cells get the right chromosomes. Science Yuncken C (1968) Meiosis in the human female. Cytogenetics
(New York) 275(5300): 632–637. 7(3): 234–238.
Page SL and Hawley RS (2004) The genetics and molecular Zickler D (2006) From early homologue recognition to synapto-
biology of the synaptonemal complex. Annual Review of Cell nemal complex formation. Chromosoma 115: 158–174.
and Developmental Biology 20: 525–558.
Perheentupa A and Huhtaniemi I (2009) Aging of the human
ovary and testis. Molecular and Cellular Endocrinology 299(1): Further Reading
2–13.
Prieto I, Tease C, Pezzi N et al. (2004) Cohesin component Alsheimer M (2009) The dance floor of meiosis: evolutionary
dynamics during meiotic prophase I in mammalian oocytes. conservation of nuclear envelope attachment and dynamics of
Chromosome Research 12: 197–213. meiotic telomeres. Genome Dynamics 5: 81–93.
Redon C, Pilch D, Rogakou E et al. (2002) Histone H2A variants de Boer E, Lhuissier FG and Heyting C (2009) Cytological
H2AX and H2AZ. Current Opinion in Genetics & Development analysis of interference in mouse meiosis. Methods in Molecular
12(2): 162–169. Biology 558: 355–382.
Rupnik A, Lowndes NF and Grenon M (2010) MRN and the race Burgoyne PS, Mahadevaiah SK and Turner JM (2009) The
to the break. Chromosoma 119: 115–135. consequences of asynapsis for mammalian meiosis. Nature
Saadallah N and Hultén MA (1986) EM investigations of surface Reviews. Genetics 10(3): 207–216.
spread synaptonemal complexes in human male carrier of a Handel MA and Schimenti JC (2010) Genetics of mammalian
pericentric inversion inv(13)(p12q14): the role of hetero- meiosis: regulation, dynamics and impact on fertility. Nature
synapsis for spermatocyte survival. Annals of Human Genetics 4: Reviews of Genetics 11: 124–136.
369–383. Harrison CJ, Alvey E and Henderson IR (2010) Meiosis in
Sharp AJ (2009) Engineering themes and new challenges in flowering plants and other green organisms. Journal of
defining the role of structural variation in human disease. Experimental Botany 61: 2863–2875.
Human Mutation 2: 135–144. Inagaki A, Schoenmakers S and Baarends WM (2010) DNA
Stahl FW and Housworth EA (2009) Methods for analysis of double strand break repair, chromosome synapsis and
crossover interference in Saccharomyces cerevisiae. Methods in transcriptional silencing in meiosis. Epigenetics 5: 255–266.
Molecular Biology 557: 35–53. Khil PP and Camerini Otero RD (2009) Variation in patterns of
Stankiewics P and Lupski JR (2010) Structural variation in the human meiotic recombination. Genome Dynamics 5: 117–127.
human genome and its role in disease. Annual Review of Medi- Neale MJ and Keeney S (2006) Clarifying the mechanics of
cine 61: 437–455. DNA strand exchange in meiotic recombination. Nature 442:
Suja JA and Barbero JL (2009) Cohesin complexes and sister 153–158.
chromatid cohesion in mammalian meiosis. Genome Dynamics Sasaki M, Lange J and Keeney S (2010) Genome destabilization
5: 94–116. by homologous recombination in the germ line. Nature Reviews
Sun F, Oliver-Bonet M, Liehr T et al. (2004) Human male in Molecular Cell Biology 11: 182–195.
recombination maps for individual chromosomes. American Sherthan H (2009) Analysis of telomere dynamics in mouse
Journal of Human Genetics 74: 521–531. spermatogenesis. Methods in Molecular Biology 558: 383–399.
Sung P and Klein H (2006) Mechanism of homologous recom- Susiarjo M, Rubio C and Hunt P (2009) Analyzing mammalian
bination: mediators and helicases take on regulatory functions. female meiosis. Methods in Molecular Biology 558: 339–335.
Nature Reviews. Molecular Cell Biology 7: 739–750. Yang F and Wang PJ (2009) The mammalian synaptonemal
Tease C, Hartshorne G and Hultén M (2006) Altered patterns of complex: a scaffold and beyond. Genome Dynamics 5: 69–80.
meiotic recombination in human fetal oocytes with asynapsis Yin S, Sun XF, Schatten H and Sun QY (2008) Molecular insights
and/or synaptonemal complex fragmentation at pachytene. into mechanisms regulating faithful chromosome separation in
Reproductive Biomedicine Online 13(1): 88–95. female meiosis. Cell Cycle 7(19): 2997–3005.

14 ENCYCLOPEDIA OF LIFE SCIENCES & 2010, John Wiley & Sons, Ltd. www.els.net

View publication stats

You might also like