ABSTRACT

BACKGROUND

The unbalanced transmission of chromosomes in human gametes and early preimplantation embryos causes aneuploidy, which is a major cause of infertility and pregnancy failure. A baseline of 20% of human oocytes are estimated to be aneuploid and this increases exponentially from 30 to 35 years, reaching on average 80% by 42 years. As a result, reproductive senescence in human females is predominantly determined by the accelerated decline in genetic quality of oocytes from 30 years of age.

OBJECTIVE AND RATIONALE

Understanding mechanisms of chromosome segregation and aneuploidies in the female germline is a crucial step towards the development of new diagnostic approaches and, possibly, for the development of therapeutic targets and molecules. Here, we have reviewed emerging mechanisms that may drive human aneuploidy, in particular the maternal age effect.

SEARCH METHODS

We conducted a systematic search in PubMed Central of the primary literature from 1990 through 2016 following the PRISMA guidelines, using MeSH terms related to human aneuploidy. For model organism research, we conducted a literature review based on references in human oocytes manuscripts and general reviews related to chromosome segregation in meiosis and mitosis.

OUTCOMES

Advances in genomic and imaging technologies are allowing unprecedented insight into chromosome segregation in human oocytes. This includes the identification of a novel chromosome segregation error, termed reverse segregation, as well as sister kinetochore configurations that were not predicted based on murine models.

WIDER IMPLICATIONS

Elucidation of mechanisms that result in errors in chromosome segregation in meiosis may lead to therapeutic developments that could improve reproductive outcomes by reducing aneuploidy.

Introduction

Human conceptions are afflicted by an extraordinary rate of chromosome errors, and the majority derive from the oocyte (Hassold and Hunt, 2001). In natural conceptions that reach clinical recognition, 35% of human pregnancies are aneuploid. The rate observed in preimplantation embryos is substantially higher, in part because aneuploid embryos have poor developmental potential and are selected against during the peri-implantation stages and throughout foetal life (Capalbo et al., 2014). In natural conception, more than 90% are of meiotic origin and the majority are caused by errors in meiosis I (Hassold and Hunt, 2001; Gabriel et al., 2011). In reproductive aged women, 20–30% of occytes (and up to 70% of oocytes in advanced maternal age (AMA) women) are aneuploid, while just 1–8% of spermatozoa are afflicted (Lu et al., 2012; Wang et al., 2012). In sperm, the incidence of aneuploidy is independent of paternal age (Erickson, 1978; Hassold and Hunt, 2001; Lu et al., 2012; Wang et al., 2012). The analyses of aneuploidy in miscarriages have been invaluable for our appreciation of the serious consequences chromosomal imbalances have for embryonic and foetal development, since a much higher incidence and wider range and representation of chromosomes are detected compared to subsequent developmental stages, including live births (Hassold et al., 1980; Zaragoza et al., 1994).

Since the discovery that aneuploidy is the major cause of congenital disorders (Jacobs and Strong, 1959; Jacobs et al., 1959; Lejeune, Gautier, and Turpin, 1959; Ford et al., 1959a,b), most our knowledge has derived from population-based studies of foetal losses and rare live births. Maternal age is the major factor that influences aneuploidy, giving rise to the characteristic J curve (Erickson, 1978; Hassold and Hunt, 2001; Fig. 1A). However, individual chromosomes follow different age-dependent curves (Nagaoka et al., 2012; Franasiak et al., 2014a, b; Fig. 1B) suggesting that both chromosome-specific as well as general cellular factors conspire to shape the segregation efficiency in human oocytes.

Aneuploidy rates in preimplantation embryos. Data from trophectoderm biopsies of human blastocysts. The data were published previously (Capalbo et al., 2013, 2014, 2015, 2016) but are presented differently for illustrative purposes (see Methods). (A) Incidence of aneuploidies in blastocysts per maternal age at oocyte retrieval. (B) Chromosome-specific incidence of aneuploidies per maternal age at oocyte retrieval. (C) Chromosome-specific proportion of monosomies and trisomies.
Figure 1

Aneuploidy rates in preimplantation embryos. Data from trophectoderm biopsies of human blastocysts. The data were published previously (Capalbo et al., 2013, 2014, 2015, 2016) but are presented differently for illustrative purposes (see Methods). (A) Incidence of aneuploidies in blastocysts per maternal age at oocyte retrieval. (B) Chromosome-specific incidence of aneuploidies per maternal age at oocyte retrieval. (C) Chromosome-specific proportion of monosomies and trisomies.

Aberrant recombination patterns on chromosomes that have mis-segregated have also been identified as an important factor, in both male and female gametes (Table I). This is because recombination together with cohesion of sister chromatids establish the unique ‘bivalent’ chromosome structure where homologous partner chromosomes are tethered together, a configuration that is critical for their accurate segregation in meiosis I (Fig. 2A). The remarkable feature is that recombination occurs in foetal oocytes whereas chromosome segregation takes place decades later (Fig. 2A). Since mammalian oocytes are arrested at the G2/M transition (or dictyate stage), this raises the intriguing question of how the bivalent is maintained until the meiotic divisions.

Table I

Mechanisms implicated in aneuploidy in human oocytes.

Function-phenotypeGene conserved?Mechanism conserved?In vivo mouse modelRefs.
RecombinationAberrant position and levelsNAYesYes(Reviewed in Nagaoka et al., 2012; Hou et al., 2013; Ottolini et al., 2015)
Age-related increase in incidence in univalentsUnivalentsNAYesNA(Henderson and Edwards, 1968; Angell, 1995; Zielinska et al., 2015)
Univalent missegregationIncreased risk of sister kinetochore splitting in meiosis I and meiosis I non-disjunctionNAYesYes(Zielinska et al., 2015; Kouznetsova et al., 2007; LeMaire-Adkins and Hunt, 2000; Koehler et al., 2006)
ShugoshinAge-related depletion in mouse oocytes (mSGO2)YesNot known; hSGO1 implicated (Garcia-Cruz, Brieno, et al., 2010a; Zielinska et al., 2015)Yes(Lister et al., 2010)
Loss of centromeric cohesionIncreased distance between sister kinetochoresNAYesYes(Duncan et al., 2012; Chiang et al., 2010; Lister et al., 2010; Zielinska et al., 2015)
MEIKINCo-orientation and centromeric cohesion in metaphase IYesYesYes(Kim et al., 2015)
SMC1βAge-related loss of bivalents; recombination normalYesNot knownYes(Revenkova et al., 2004; Hodges et al., 2005; Murdoch et al., 2013)
HDAC activity & chromosome compactionDecreased deacetylation of chromosomesYesYesYes(van den Berg et al., 2011)
Spindle instabilityUnstable spindles in human oocytes age-independentNAHuman specificNA(Holubcova et al., 2015)
SAC- Aurk B and CVariants associated with aneuploidy in women at IVF clinicsYesNot known (section on tension-mediated segregation)No(Nguyen et al., 2017)
Function-phenotypeGene conserved?Mechanism conserved?In vivo mouse modelRefs.
RecombinationAberrant position and levelsNAYesYes(Reviewed in Nagaoka et al., 2012; Hou et al., 2013; Ottolini et al., 2015)
Age-related increase in incidence in univalentsUnivalentsNAYesNA(Henderson and Edwards, 1968; Angell, 1995; Zielinska et al., 2015)
Univalent missegregationIncreased risk of sister kinetochore splitting in meiosis I and meiosis I non-disjunctionNAYesYes(Zielinska et al., 2015; Kouznetsova et al., 2007; LeMaire-Adkins and Hunt, 2000; Koehler et al., 2006)
ShugoshinAge-related depletion in mouse oocytes (mSGO2)YesNot known; hSGO1 implicated (Garcia-Cruz, Brieno, et al., 2010a; Zielinska et al., 2015)Yes(Lister et al., 2010)
Loss of centromeric cohesionIncreased distance between sister kinetochoresNAYesYes(Duncan et al., 2012; Chiang et al., 2010; Lister et al., 2010; Zielinska et al., 2015)
MEIKINCo-orientation and centromeric cohesion in metaphase IYesYesYes(Kim et al., 2015)
SMC1βAge-related loss of bivalents; recombination normalYesNot knownYes(Revenkova et al., 2004; Hodges et al., 2005; Murdoch et al., 2013)
HDAC activity & chromosome compactionDecreased deacetylation of chromosomesYesYesYes(van den Berg et al., 2011)
Spindle instabilityUnstable spindles in human oocytes age-independentNAHuman specificNA(Holubcova et al., 2015)
SAC- Aurk B and CVariants associated with aneuploidy in women at IVF clinicsYesNot known (section on tension-mediated segregation)No(Nguyen et al., 2017)

This list is not exhaustive but contains references to some of the seminal studies that may be directly relevant to human aneuploidy. SAC, spindle assembly checkpoint; HDAC, histone deacetylase; univalents: single chromosomes without physical connections to their partner (homologue).

Table I

Mechanisms implicated in aneuploidy in human oocytes.

Function-phenotypeGene conserved?Mechanism conserved?In vivo mouse modelRefs.
RecombinationAberrant position and levelsNAYesYes(Reviewed in Nagaoka et al., 2012; Hou et al., 2013; Ottolini et al., 2015)
Age-related increase in incidence in univalentsUnivalentsNAYesNA(Henderson and Edwards, 1968; Angell, 1995; Zielinska et al., 2015)
Univalent missegregationIncreased risk of sister kinetochore splitting in meiosis I and meiosis I non-disjunctionNAYesYes(Zielinska et al., 2015; Kouznetsova et al., 2007; LeMaire-Adkins and Hunt, 2000; Koehler et al., 2006)
ShugoshinAge-related depletion in mouse oocytes (mSGO2)YesNot known; hSGO1 implicated (Garcia-Cruz, Brieno, et al., 2010a; Zielinska et al., 2015)Yes(Lister et al., 2010)
Loss of centromeric cohesionIncreased distance between sister kinetochoresNAYesYes(Duncan et al., 2012; Chiang et al., 2010; Lister et al., 2010; Zielinska et al., 2015)
MEIKINCo-orientation and centromeric cohesion in metaphase IYesYesYes(Kim et al., 2015)
SMC1βAge-related loss of bivalents; recombination normalYesNot knownYes(Revenkova et al., 2004; Hodges et al., 2005; Murdoch et al., 2013)
HDAC activity & chromosome compactionDecreased deacetylation of chromosomesYesYesYes(van den Berg et al., 2011)
Spindle instabilityUnstable spindles in human oocytes age-independentNAHuman specificNA(Holubcova et al., 2015)
SAC- Aurk B and CVariants associated with aneuploidy in women at IVF clinicsYesNot known (section on tension-mediated segregation)No(Nguyen et al., 2017)
Function-phenotypeGene conserved?Mechanism conserved?In vivo mouse modelRefs.
RecombinationAberrant position and levelsNAYesYes(Reviewed in Nagaoka et al., 2012; Hou et al., 2013; Ottolini et al., 2015)
Age-related increase in incidence in univalentsUnivalentsNAYesNA(Henderson and Edwards, 1968; Angell, 1995; Zielinska et al., 2015)
Univalent missegregationIncreased risk of sister kinetochore splitting in meiosis I and meiosis I non-disjunctionNAYesYes(Zielinska et al., 2015; Kouznetsova et al., 2007; LeMaire-Adkins and Hunt, 2000; Koehler et al., 2006)
ShugoshinAge-related depletion in mouse oocytes (mSGO2)YesNot known; hSGO1 implicated (Garcia-Cruz, Brieno, et al., 2010a; Zielinska et al., 2015)Yes(Lister et al., 2010)
Loss of centromeric cohesionIncreased distance between sister kinetochoresNAYesYes(Duncan et al., 2012; Chiang et al., 2010; Lister et al., 2010; Zielinska et al., 2015)
MEIKINCo-orientation and centromeric cohesion in metaphase IYesYesYes(Kim et al., 2015)
SMC1βAge-related loss of bivalents; recombination normalYesNot knownYes(Revenkova et al., 2004; Hodges et al., 2005; Murdoch et al., 2013)
HDAC activity & chromosome compactionDecreased deacetylation of chromosomesYesYesYes(van den Berg et al., 2011)
Spindle instabilityUnstable spindles in human oocytes age-independentNAHuman specificNA(Holubcova et al., 2015)
SAC- Aurk B and CVariants associated with aneuploidy in women at IVF clinicsYesNot known (section on tension-mediated segregation)No(Nguyen et al., 2017)

This list is not exhaustive but contains references to some of the seminal studies that may be directly relevant to human aneuploidy. SAC, spindle assembly checkpoint; HDAC, histone deacetylase; univalents: single chromosomes without physical connections to their partner (homologue).

Chromosome recombination and segregation in human oocytes. (A) Homologous chromosomes recombine (Rec) during meiotic prophase I in foetal oocytes. The cohesion between the sister chromatids holds the resulting X-shape (chiasma) in place. This configuration is known as the ‘bivalent’ and is only seen physically once chromosomes compact in metaphase I. Until menarche, the oocyte is arrested with diffuse chromatin, known as dictyate arrest. Upon ovulation and resumption of meiosis in fully grown oocytes, the nuclear envelope (germinal vesicle, GV) breaks down and metaphase I ensues. Chromosomes compact and segregate at anaphase I, resulting in the extrusion of one of the homologues in the first polar body (PB1). Because recombination is suppressed near centromeres, the pericentromeric sequences, illustrated in blue and red, can be used to ‘fingerprint’ chromosomes and determine segregation patterns. (B) The types of segregation in meiosis I and meiosis II are shown. The error rates (% of chromosomes) for each type of pattern, which matches the colour below the segregation pattern to the right, is illustrated in the graph. The data are shown from Hou et al. (2013), who used the female pronucleus (FPN) obtained from oocytes from young women, whereas the data from artificially-activated oocytes or embryos are from Ottolini et al. (2015), who used oocytes from women of AMA. (C) MeioMapping consists of determining genome-wide recombination and segregation patterns. Mature MII oocytes are biopsied (PB1) followed by artificial activation, allowing the recovery of the PB2 and the oocyte. (D) The DNA of the three cells is amplified, SNPs are detected and phased and this is followed by mapping of reciprocal haplotype blocks and recombination sites from which the ‘bivalent’ configuration can be inferred.
Figure 2

Chromosome recombination and segregation in human oocytes. (A) Homologous chromosomes recombine (Rec) during meiotic prophase I in foetal oocytes. The cohesion between the sister chromatids holds the resulting X-shape (chiasma) in place. This configuration is known as the ‘bivalent’ and is only seen physically once chromosomes compact in metaphase I. Until menarche, the oocyte is arrested with diffuse chromatin, known as dictyate arrest. Upon ovulation and resumption of meiosis in fully grown oocytes, the nuclear envelope (germinal vesicle, GV) breaks down and metaphase I ensues. Chromosomes compact and segregate at anaphase I, resulting in the extrusion of one of the homologues in the first polar body (PB1). Because recombination is suppressed near centromeres, the pericentromeric sequences, illustrated in blue and red, can be used to ‘fingerprint’ chromosomes and determine segregation patterns. (B) The types of segregation in meiosis I and meiosis II are shown. The error rates (% of chromosomes) for each type of pattern, which matches the colour below the segregation pattern to the right, is illustrated in the graph. The data are shown from Hou et al. (2013), who used the female pronucleus (FPN) obtained from oocytes from young women, whereas the data from artificially-activated oocytes or embryos are from Ottolini et al. (2015), who used oocytes from women of AMA. (C) MeioMapping consists of determining genome-wide recombination and segregation patterns. Mature MII oocytes are biopsied (PB1) followed by artificial activation, allowing the recovery of the PB2 and the oocyte. (D) The DNA of the three cells is amplified, SNPs are detected and phased and this is followed by mapping of reciprocal haplotype blocks and recombination sites from which the ‘bivalent’ configuration can be inferred.

Here, we review recent technological breakthroughs in single-cell genomics and time-lapse imaging of human oocytes, which are revealing unexpected new facets of chromosome segregation. In particular, we discuss the emerging model of Chromosomal Aging, which integrates and relies upon knowledge from human and other mammalian oocytes to explain age-related aspects of female aneuploidy. New insights into the spindle assembly checkpoint (SAC) and spindle formation in human oocytes reveal that the spindle itself may be error-prone. Advances in technology and in vitro biology usher in the potential for novel therapies that could extend the reproductive lifespan of women (Andersen, 2015).

Methods

To evaluate molecular mechanisms that have been implicated in human female meiosis, we carried out a systematic review following the PRISMA guidelines. Pubmed Central was searched for the following MeSH terms: chromosome segregation, human embryo, human oocyte, human germline, meiosis, aneuploidy, meiotic disturbances, meiotic protective factors and aneuploidy, for all full-text articles published in English from 1990 through 2016. Keywords were used in multiple and overlapping combinations to identify those publications strictly relevant to oocyte meiosis. This identified 29 213 publications. Of these, 3855 were reviews and excluded. The remaining abstracts were screened for whether human oocytes or embryos were included. This eliminated 12 953 articles. The remaining 12 405 abstracts were screened further for the inclusion of ‘human aneuploidy’ or ‘human meiosis’. This reduced the number of relevant publications to 295, which were read in full. Reference lists were also cross-checked for additional relevant studies.

For the models for chromosome segregation in meiosis, no systematic review was carried out. We used the references lists from human oocyte studies and relied on reviews from the meiosis field to identify relevant mechanistic studies. The animal studies therefore do not represent a comprehensive review of all available models for aneuploidy.

For illustrative purposes, embryo aneuploidy data from previous publications were retrieved and elaborated. Aneuploidy data were obtained by means of array (a) CGH or quantitiative (q) PCR-based 24-chromosome testing (Capalbo et al., 2013, 2014, 2015, 2016). Whole chromosome aneuploidies were stratified according to female age used as continuous variable (per each year unit) to report chromosome-specific susceptibility to oocyte aging. Data were also stratified at the single chromosome level to investigate monosomy/trisomy ratio observed in embryos. Overall, data from approximately 5000 embryos were included in this elaboration. Only a descriptive examination of peer-reviewed data is reported here for illustrative purposes and without additional inferential analysis performed.

Results

A new genomics era for single cells: recovery of genetic information from all products of meiosis

We have long appreciated the power of being able to detect both the genetic information and chromosomal content in all products of meiosis to infer chromosome segregation patterns and genetic inheritance (Fig. 2A). Specifically, obtaining the recombination patterns allows ‘reconstruction’ of what bivalents might have looked like, prior to chromosome segregation in the meiotic divisions (Fig. 2C). Such analyses, recently been developed for human oocytes and their matched polar bodies, are referred to as MeioMapping (Fig. 2B and C) (Ottolini et al., 2015). Two independent groups sequentially biopsied the polar bodies by either activating the oocyte artificially (Ottolini et al., 2015) or fertilizing the mature oocyte using sperm (Hou et al., 2013; Ottolini et al., 2015). After fertilization, the female pronucleus was extracted (Hou et al., 2013) or a biopsy of the blastocyst was obtained (Ottolini et al., 2015). This is followed by amplification of the DNA, termed whole-genome amplification (WGA; Zhang et al., 1992), which yields sufficient quantity of DNA to create libraries for next-generation sequencing (NGS) or single-nucleotide polymorphism (SNP) arrays (Fig. 3). WGA was initially applied to screen polar bodies for aneuploidies using a content analysis known as comparative genomic hybridization (CGH; Wells et al., 2002). SNP arrays or NGS allow not only content but also genetic information to be discerned (Fig. 3).

Having the known parental haplotypes (KPH) allows phasing and inferences about chromosome segregation and recombination. (A) Content analysis versus (B) KPH. (C–E) Content analyses includes fluorescence in-situ hybridization (FISH), array comparative genomic hybridization (CGH), and SNP array/next-generation sequencing (NGS) without phasing. (F) Illustraion of haplotype detection in the three cells of a single meioses when the phase (red and blue) is known.
Figure 3

Having the known parental haplotypes (KPH) allows phasing and inferences about chromosome segregation and recombination. (A) Content analysis versus (B) KPH. (CE) Content analyses includes fluorescence in-situ hybridization (FISH), array comparative genomic hybridization (CGH), and SNP array/next-generation sequencing (NGS) without phasing. (F) Illustraion of haplotype detection in the three cells of a single meioses when the phase (red and blue) is known.

Several important observations have been made using MeioMapping because it allows the recovery of all chromosomes involved in the meiotic divisions. Using the pericentromeric SNPs for chromosome fingerprinting, Ottolini and colleagues discovered a new segregation pattern, termed reverse segregation (Fig. 2A). Reverse segregation is characterized by both homologous chromosome segregating their sister chromatids at meiosis I. At meiosis II, the two non-sister chromatids segregate correctly in nearly 80% of cases (Ottolini et al., 2015).

MeioMapping has revealed that although the major aberrant segregation defect is reverse segregation in oocytes from women of AMA (Ottolini et al., 2015), predivision or precocious separation of sister chromatids (PSSC) causes the majority of aneuploidy in activated/fertilized oocytes (Hou et al., 2013; Ottolini et al., 2015). This is because non-sister chromatids segregate to opposite spindle poles with relatively high efficiency at meiosis II during reverse segregation (Ottolini et al., 2015). The molecular mechanisms are unknown. MeioMapping has shown that ~70% of errors occur in meiosis I, but only half of these cause aneuploidy in the oocyte, since ‘corrective’ segregation events at meiosis II may compensate for the initial error (Hou et al., 2013; Ottolini et al., 2015). This is consistent with findings from content analyses of polar bodies and embryos (Verlinsky et al., 2001; Pellestor et al., 2002, 2005; Fragouli et al., 2009; Handyside et al., 2012; Capalbo et al., 2013; Forman et al., 2013). All of the chromosome segregation errors observed in the two studies that followed the chromosomes with fingerprinting also firmly established that the segregation errors are reciprocal, affecting two of the three cells, and therefore arose during meiosis. This is also consistent with findings from chromosomal content analyses (Verlinsky et al., 2001; Pellestor et al., 2002, 2005; Fragouli et al., 2009; Handyside et al., 2012; Capalbo et al., 2013) and is important clinically, since polar bodies can be used to infer aneuploidy in oocytes. This has been the basis for preimplantation genetic testing for aneuploidy (PGT-A; Fig. 4).

Human female meiosis and preimplantation genetic testing for aneuploidy (PGT-A).
Figure 4

Human female meiosis and preimplantation genetic testing for aneuploidy (PGT-A).

Seeing is believing: time-lapse imaging of chromosomes in real time reveals splitting of sister kinetochores at meiosis I

The inference that human chromosomes can split their sister chromatids at meiosis I, during reverse segregation or a single predivision (PSSC) should perhaps not have come as a surprise. The single X chromosome in XO mouse oocytes frequently segregate sister chromatids at meiosis I (LeMaire-Adkins and Hunt, 2000). Development of time-lapse imaging allowed the first visualization of sister chromatid splitting at meiosis I. This came from mouse oocytes deleted for Sycp3 (Kouznetsova et al., 2007), a synaptonemal complex gene that promotes crossing over (Yuan et al., 1998, 2000). In oocytes of Sypc3-/- females, a few homologue pairs fail to generate crossovers and the chromosomes remain univalent. The univalents segregate both as whole chromosomes or split their sister chromatids at meiosis I (Kouznetsova et al., 2007; Sakakibara et al., 2015).

Direct observation that human chromosomes can split their sister chromatids at meiosis I came when fluorescence time-lapse imaging of human oocytes was achieved. Human immature GV oocytes were injected with mRNA encoding fluorescently labelled kinetochores and spindle proteins, which allowed Zielinska and colleagues to follow the fate of chromosomes during the first meiotic division (Zielinska et al., 2015). The incidence of sister chromatid splitting increased with maternal age. The splitting of sister chromatids in meiosis I provided direct evidence for both the reverse segregation and PSSC patterns that have been inferred from genetic data (Hou et al., 2013; Ottolini et al., 2015) and cytological observations of fixed material (Angell, 1991; Fig. 5B). Notably, human MII oocytes also contain whole chromosome gains and losses (Jagiello et al., 1975; Polani and Jagiello, 1976; Martin et al., 1986; Pellestor et al., 2002), suggesting that PSSC and reverse segregation co-exist with meiosis I non-disjunction. It is currently unclear what factors determine their relative frequencies in oocytes obtained from IVF clinics.

Modifications to meiotic chromosomes that promote homologous chromosome segregation in the first division. (A) These two modifications to meiotic chromosomes prior to the first division can be regulated separately, as seen in some organisms where different complexes mediate co-orientation and cohesion at sister centromeres, respectively (Petronczki et al., 2003). In mammalian oocytes, the conserved MEIKEN mediates both (Kim et al., 2015). Furthermore, in murine oocytes Shugoshin (mSgo2, Japanese for ‘guardian spirit’,) protects centromeric cohesin from degradation at meiosis I (Lee et al., 2008), such that sister chromatids are held together until the second division. Depleted protein levels of mSgo2 are associated with loss of centromeric cohesion and with elevated levels of aneuploidy in aged murine oocytes (Lister et al., 2010). Cohesion loss at centromeres has also been seen in a second study of naturally-aged mice (Chiang et al., 2010) as well as in oocytes from women of advantaged maternal age (Duncan et al., 2012; Zielinska et al., 2015). (B) Example of spread and fixed chromosomes from a human metaphase I oocyte. Kinetochores are shown in magenta and chromosomes in grey. The pair of univalent (green circle) has been enlarged in the green box (left hand corner). Scale bar: 5 μm. (C) Univalent formation increases the risk that sister kinetochores segregation to opposite spindle poles in meiosis I, resulting in reverse segregation. (D) Bivalent configurations where cohesion loss near centromeres generate half or fully-inverted configurations that may precede PSSC or reverse segregation, respectively. (E) Spindle instability with multiple spindles (left) may result in sister kinetochores being attached to microtubules from opposite spindle poles once the bipolar spindle is formed (right hand side).
Figure 5

Modifications to meiotic chromosomes that promote homologous chromosome segregation in the first division. (A) These two modifications to meiotic chromosomes prior to the first division can be regulated separately, as seen in some organisms where different complexes mediate co-orientation and cohesion at sister centromeres, respectively (Petronczki et al., 2003). In mammalian oocytes, the conserved MEIKEN mediates both (Kim et al., 2015). Furthermore, in murine oocytes Shugoshin (mSgo2, Japanese for ‘guardian spirit’,) protects centromeric cohesin from degradation at meiosis I (Lee et al., 2008), such that sister chromatids are held together until the second division. Depleted protein levels of mSgo2 are associated with loss of centromeric cohesion and with elevated levels of aneuploidy in aged murine oocytes (Lister et al., 2010). Cohesion loss at centromeres has also been seen in a second study of naturally-aged mice (Chiang et al., 2010) as well as in oocytes from women of advantaged maternal age (Duncan et al., 2012; Zielinska et al., 2015). (B) Example of spread and fixed chromosomes from a human metaphase I oocyte. Kinetochores are shown in magenta and chromosomes in grey. The pair of univalent (green circle) has been enlarged in the green box (left hand corner). Scale bar: 5 μm. (C) Univalent formation increases the risk that sister kinetochores segregation to opposite spindle poles in meiosis I, resulting in reverse segregation. (D) Bivalent configurations where cohesion loss near centromeres generate half or fully-inverted configurations that may precede PSSC or reverse segregation, respectively. (E) Spindle instability with multiple spindles (left) may result in sister kinetochores being attached to microtubules from opposite spindle poles once the bipolar spindle is formed (right hand side).

Two different mechanisms appear to be involved in sister chromatid splitting in human oocytes (Zielinska et al., 2015). Univalents are sometimes apparent prior to sister chromatid splitting (Fig. 5C). Univalents or individual sister chromatids have been seen previously in human MI oocytes (Angell, 1995; Garcia-Cruz et al., 2010a, b; Fig. 5B) and their incidence is age-related in both mouse (Henderson and Edwards, 1968) and human (Angell, 1995; Zielinska et al., 2015). Although there is extensive evidence that this is due to age-related deterioration in cohesion in mouse oocytes, the mechanisms for their age-related incidence in human oocytes is still not clear, as discussed below. In other cases, the bivalent still appeared to be intact, but one or both the chromosomes still oriented their sister kinetochores in opposite directions, termed ‘half-inverted’ or ‘fully-inverted’ configurations (Fig. 5D).

Spindle instability and unfused sister kinetochores are permissive of erroneous kinetochore-microtubule attachments

What may cause sister kinetochores to separate precociously? Human meiotic spindles are not only different in their architecture but highly unstable compared to those found in mouse oocytes (Holubcova et al., 2015). Occurring without centrosomes (acentrosomal), spindle formation initiates from chromosomes and the spindles take up to five hours to form after breakdown of the nuclear envelope (germinal vesicle break down). Eight of out ten spindles become multipolar prior to taking on a bipolar configuration. Metaphase I takes an extremely long time, 14–16 h (Angell, 1995; Holubcova et al., 2015) and although the spindle takes on a bipolar configuration prior to the onset of anaphase I and division, the severity of instability is correlated with attachment of sister kinetochores to opposite spindle poles (Fig. 5E). Spindle instability was reported to be age-independent (Holubcova et al., 2015), a finding in contrast with previous studies of fixed oocytes (Battaglia et al., 1996), including oocytes from naturally cycling women.

Spindle instability alone, however, does not explain why sister kinetochores bi-orient. Sister kinetochores are normally co-orientated and sister chromatids are topologically connected by catenation of the sister DNA molecules and protein-mediated cohesion (cohesin complexes; Fig. 5A). In many organisms, sister kinetochores are fused, however, in human oocytes this may not be the case (Patel et al., 2015). Instead, sister kinetochores are often attached to spindle fibres, known as k-fibres, from opposite spindle poles (Patel et al., 2015; Zielinska et al., 2015). The incidence of sister kinetochore splitting and, more generally, the distance between sister kinetochores (termed iKT distances), have been reported to correlate with increased maternal age in both mice (Chiang et al., 2010; Lister et al., 2010) and humans (Duncan et al., 2012; Patel et al., 2015; Zielinska et al., 2015; Lagirand-Cantaloube et al., 2017). The impact on chromosome segregation as determined by the physical distances between sister kinetochores remain somewhat unclear.

The chromosomal aging hypothesis: cohesion loss precedes chromosome segregation errors

As discussed above, the incidence of univalents and sister kinetochore splitting in meiosis I is age-related in both mouse and human oocytes. There is also evidence that this leads to elevated risk of chromosome missegregation in meiosis I or meiosis II (Koehler et al., 2006; Kouznetsova et al., 2007; Chiang et al., 2010; Lister et al., 2010; Merriman et al., 2013; Sakakibara et al., 2015; Zielinska et al., 2015). What is less clear, however, is the origin of univalents and their age-related incidence, which was first noted by Henderson and Edwards (1968) for mouse oocytes. In their seminal study, the authors attempted to reconcile findings from population studies that aneuploidies were often associated with ‘crossover-less’ recombination patterns. They proposed two not mutually exclusive hypotheses: (i) that univalents originated from bivalents that deteriorated during dictyate arrest; or (ii) that univalent incidence increased with maternal age because oocytes entered and exited meiosis (ovulation) in waves, such that foetal oocytes in the early waves had ‘good’ recombination and subsequently good segregation, when females were young butas females aged, oocytes originating from ‘later’ foetal waves would be ovulated, and these had poorer segregation because the recombination was worse. This latter hypothesis is also known as the Production Line hypothesis.

The first part of the Production Line hypothesis predicts that recombination rates should follow a gradient in foetal oocytes, however, this is not the case (Rowsey et al., 2014). The second part, that oocytes with lower recombination rates and therefore compromised segregation potential ovulate later, has not been tested. If anything, recombination rates in children increase as women age (Kong et al., 2004; Coop et al., 2008; Campbell et al., 2015), although this can also be explained by the selection for oocytes with high recombination rates and therefore lower aneuploidy risk (Ottolini et al., 2015). There are several scientific observations that makes it difficult to rule out ovulation timing. Unlike mouse oocytes, human foetal and adult oocytes show tremendous heterogeneity in their recombination rates, in some cases by an order of magnitude (Lenzi et al., 2005; Hou et al., 2013; Ottolini et al., 2015). The incidence of crossover-less foetal oocytes is also extremely high, reported to be nearly 20% in some studies (Tease et al., 2002; Cheng et al., 2009). Thus, preferential ovulation timing could explain the high incidence of univalents in oocytes from women of AMA. Such a model requires that (i) cell-cycle progression over a time scale of decades is linked to recombination status; and (ii) follicular response is dependent upon the cell-cycle status of the oocyte. Although these two requirements are possible, since the meiotic silencing checkpoint determines progression in response to synapsis and recombination (Turner, 2015) and oocyte attrition due to DNA damage halts follicular development (Titus et al., 2013), they remain to be tested or shown to be important for aneuploidy generation in human.

There is ample evidence that cohesion loss may drive the age-dependent univalent formation in mouse. The most conclusive is from the Smc1β-/- female mouse where recombination levels are narrowly distributed in foetal oocytes, but the incidence of univalents (and aneuploidy) follows an age-dependent gradient (Hodges et al., 2005). SMC1β is particularly intriguing because it is part of the meiotic cohesin complex that mediates cohesion (Fig. 5A) and is haploinsufficient for aneuploidy (Murdoch et al., 2013). In general, because mouse oocytes show highly regulated recombination with virtually no crossover-less oocytes (Hunt and Hassold, 2008), the majority of age-related effects on chromosome structure can be attributed to cohesion loss. Age-dependent cohesion loss accounts for both univalent formation and the loss of sister chromatid cohesion near centromeres. Hence, cohesion deterioration or exhaustion provides a single molecular mechanism for the loss of bivalent maintenance as females age (Jessberger, 2012).

One important prediction of the chromosomal aging model is that if cohesin loss is the cause of aneuploidy, then bivalent deterioration should be correlated to cohesin loss or dysregulation. This appears to be the case in mouse MI oocytes where mSGO2 and mREC8 (Fig. 5A) are preferentially lost from centromeres where the two sister kinetochores are split (Lister et al., 2010). In human oocytes, this prediction is not upheld. Although univalents are observed with increased incidence in oocytes from women of AMA (Henderson and Edwards, 1968; Angell, 1995; Zielinska et al., 2015), univalent structures were not preferentially depleted for cohesin (REC8) compared to bivalent chromosomes in oocytes from women 35 years or younger (Garcia-Cruz, et al., 2010a). Thus, although other studies have reported diminished levels of cohesin proteins (total levels) and mRNAs (Tsutsumi et al., 2014), the chromosomal association of cohesin does not correlate with bivalent structures, at least according to the results of this one study (Garcia-Cruz et al., 2010a). However, it is possible that cohesion maintenance is affected, since only a proportion of cohesin complexes mediate cohesion in mitotic cells (Nishiyama et al., 2010). Having an underlying mechanism for cohesion loss in human oocytes is clearly needed.

The cohesion exhaustion hypothesis is only one of several mechanisms by which meiotic chromosomes may age or be aberrant (age-independent). Defective deacetylation of histone H4 K12 is seen with greater incidence in aged oocytes (human) and has been suggested to account for compaction and chromosome alignment defects, resulting in aneuploidy (van den Berg et al., 2011). Shorter telomeres were preferentially associated with aneuploidy in a study of polar bodies and preimplantation embryos (Treff et al., 2011). A study of preimplantation embryos also reported that aneuploid embryos from women of AMA or suffering from recurrent miscarriage also had, on average, shorter telomeres (Mania et al., 2014). Telomere regulation, however, is challenging to study and changes according to developmental stage (Turner et al., 2010; Turner and Hartshorne, 2013). Telomerase, which extends telomeres, is required for chromosome alignment and spindle stability at metaphase I in mouse (Liu et al., 2002). However, we currently do not know whether this is due to telomeric effects on chromosome segregation or whether the correlation can be explained by the important function of telomeres in recombination and chromosome pairing during the preceding meiotic prophase in foetal oocytes (Shibuya and Watanabe, 2014).

Chromosome-specific maternal age curves

One intriguing feature of chromosomal aging is that it must interact with other factors that cause chromosomes to follow different age-dependent aneuploidy curves (Nagaoka et al., 2012; Franasiak et al., 2014a, b) (Fig. 1B). As illustrated in Fig. 1B, chromosomes 1–9 are relatively shallow, whereas the acrocentric chromosomes and especially 21 and 22 show high rates of missegregation. Chromosomes 15 and 16 are different yet again (Nagaoka et al., 2012).

One major factor that influences chromosome segregation and potentially also sister chromatid cohesion is recombination. The influence of recombination on aneuploidy was first discovered by studying spontaneous miscarriages once chromosome fingerprinting became available (Nagaoka et al., 2012). The positions of recombination events where homologues become tethered appeared perturbed when chromosomes had mis-segregated at meiosis I (Lamb et al., 2005). Some crossovers are ‘too close’ or ‘too far’ away from centromeres and some pairs are crossover-less. Importantly, chromosomes display specific vulnerabilities (Nagaoka et al., 2012). Further studies of recombination in the US National Down Syndrome Project identified several intriguing features, including that Trisomy 21 individuals tended to have a global decrease in recombination events compared to their unaffected siblings (Middlebrooks et al., 2013). Using the approach of mapping recombination across all three products from a single meiosis, global recombination levels were shown to correlate positively with euploidy in both younger women (Hou et al., 2013) and women of AMA (Ottolini et al., 2015). In addition, crossover-less chromosomes are at increased risk of PSSC and reverse segregation, since sister chromatids that did not participate in a recombination event are also at elevated risk of PSSC (Ottolini et al., 2015). Finally, recombination also appears to determine how sister chromatids segregate from each other at meiosis II. Recombinant sister chromatids are more likely to be retained in the oocyte compared to the second polar body in both human (Ottolini et al., 2015) and mouse (Wu et al., 2005). The mechanisms underlying these features are not known.

Pertinent to the discovery that sister chromatids are often separated at meiosis I, recombination near centromeres is associated with aneuploidy in human conceptions (Nagaoka et al., 2012). Whereas terminal crossovers may provide less rigidity of bivalents during tension-mediated attachment to microtubules (Webster and Schuh, 2016), centromeric crossovers may disrupt cohesion or the crossover may become entrapped in the centromeric domain of cohesin that is protected and maintained until meiosis II (Fig. 5A). Studies in fruitflies and budding yeast have also found that recombination near or at centromeres is detrimental for chromosomes (Koehler et al., 1996; Rockmill et al., 2006). Studying the direct link between recombination and sister chromatid dynamics with combined live cell imaging and genomics will likely yield important insight into the impact that centromeric and telomeric crossovers have on chromosome segregation.

Reconstructing the bivalent configuration from MeioMaps: recombination and its link with chromosome segregation

The combined assessment of haplotypes that are determined by recombination also allowed the first direct correlations between recombination and chromosome segregation in single meiosis (Fig. 2). Traditionally, recombination has been studied by assessing MLH1 foci, a cytological marker that correlates in numbers with chiasmata and whose deletion reduces crossover rates or abrogates meiosis in model organisms (Baker et al., 1996; Edelmann et al., 1996; Hunter and Borts, 1997; Tease et al., 2002). Studies from natural pregnancies have firmly established that recombination rates and their position along chromosomes are important in the aetiology of aneuploidy in foetuses and rare live births. Hou et al. (2013) reported that the link between recombination and errors in chromosome segregation is relatively weak in young women, whereas Ottolini et al. (2015) found that recombination frequency explains 18% of the variability in aneuploidy in oocytes obtained from women of AMA. In both cases, a lower genome-wide rate of recombination events lead to higher susceptibility to segregation errors during meiosis.

Epidemiologically, recombination and maternal age are still the strongest factors associated with the risk of aneuploidies. Recombination rates are also linked to increased reproductive success in women, especially as they age (Kong et al., 2004). It is possible that increased recombination events within an oocyte could counteract bivalent deterioration over time thus ensuring normal segregation and a euploid conception. This could explain the elevated recombination rates in children born to ‘older’ mothers in several populations (Kong et al., 2004; Coop et al., 2008; Campbell et al., 2015).

The crossover paradox

If recombination levels are important to ensure chromosome segregation, why does female meiosis generate more ‘vulnerable’ configurations including crossover-less pairs compared to male meiosis, who only have half the number of crossovers (Tease, et al., 2002; Lenzi et al., 2005; Cheng et al., 2009; Ottolini et al., 2015)? Vulnerable crossover configurations such as crossover-less pairs are more than an order of magnitude greater in foetal oocytes than sperm (Cheng et al., 2009). What causes ‘recombination failure’ on chromosome pairs in oocytes compared to adult sperm, where such configurations are virtually never seen? Recent mathematical modelling of crossover distribution in human foetal oocytes suggests that specific steps in the maturation of crossovers may be inefficient in oocytes, but not sperm (Wang et al., 2017). This could explain not only the incidence of crossover-less chromosome pairs but also other vulnerable configurations (e.g. terminal positions) that predispose chromosomes to missegregation decades later, when the adult oocyte ovulates and the meiotic divisions take place.

Do cellular checkpoints contribute to poor genetic quality of human oocytes?

MeioMapping in human oocytes revealed one catastrophic meiosis, where none of the chromosome pairs had recombined and 12 pairs had mis-segregated, leading to gross aneuploidy in the MII oocyte (Ottolini et al., 2015). In this case, a complete failure to initiate meiotic recombination could explain the apparent lack of recombination genome-wide. What is equally interesting is that such oocytes mature suggesting that the meiotic silencing checkpoint is not stringent enough to eliminate defective foetal oocytes from the adult pool (Turner, 2015). The meiotic silencing checkpoint normally eliminates oocytes or spermatocytes that fail to pair (synapse) homologous chromosomes, a process that is highly dependent upon recombination. However, the meiotic silencing checkpoint appears to be noticeably less efficient in female mice than male mice (Cloutier et al., 2015). If the same is true for human oocytes, then inefficient elimination of oocytes with aberrant or vulnerable crossover configurations generated by the highly heterogenous recombination process (Tease et al., 2002; Lenzi et al., 2005) could contribute to the high rate of aneuploidy.

The DNA damage response (DDR) mediates repair as well as cellular arrest or apoptosis in response to endogenous and exogenous damage (Jackson and Bartek, 2009). In meiosis, the DDR also induces recombination via the formation of hundreds of double-strand breaks (Keeney et al., 2014). Most DNA damage repair genes appear to be important for culling oocytes (or spermatocytes) in response to persistent damage (Bolcun-Filas et al., 2014). Mutants that result in damage that does not trigger arrest, such as recombination intermediates (Hwang et al., 2017), could contribute to aneuploidy in affected human oocytes.

The SAC controls the onset of anaphase. In mitotic cells, the SAC monitors correct kinetochore attachment and promotes re-orientation until sister kinetochores are under tension from attachment to opposite spindle poles. Only then is anaphase onset triggered and separase activated (Fig. 5A), allowing sister chromatids to separate. Unattached kinetochores trigger the accumulation of SAC components and formation of the mitotic checkpoint complex (MCC; Cdc20-BubR1, Bub3 and Mad2), which inhibits the activation of the anaphase promoting complex. The SAC is also required to prevent aneuploidy in mouse oocytes (Wassmann et al., 2003; Homer et al., 2005; Niault et al., 2007; McGuinness et al., 2009; Hached et al., 2011; Touati et al., 2015), presumably by ensuring the timely onset of anaphase, which is accelerated when Bub1 or BubR1 are depleted or when only a single copy of MAD2 is present. There is therefore substantial evidence that the SAC is critical for mammalian oocytes, but the question is whether the SAC works the same way in meiosis as in mitosis.

Several lines of evidence suggest that the mechanism that triggers the SAC might be adapted in meiosis. In mitosis, the stretching of sister kinetochores that are under tension from microtubules emanating from opposite spindle poles physically removes Aurora B kinase from its phosphorylation targets, thereby preventing de-stabilization of kinetochore-microtubule interactions, or ‘silencing’ the SAC. Recent observations in mouse oocytes question this model for bivalents in meiosis I. Both Aurora B and Aurora C are localized close to kinetochore-microtubule attachments when these are under tension, suggesting that mouse oocytes do not contain a mechanism for ‘stretching’-dependent phospho-regulation of kinetochore-microtubule attachments (Yoshida et al., 2015). The bivalent tension or stretching model originated from the elegant ‘pricking’ experiments conducted in grasshopper spermatocytes, where bivalents ceased to reorient when a needle that mimicked tension was applied (Nicklas and Koch, 1969). Tension could still be monitored in mammalian oocytes, but the mechanism for kinetochore-microtubule re-orientation would then have to be different compared to mitotic cells.

The second major finding in recent years is that the SAC responds to exogenous DNA damaging agents by arresting cells in MI (Collins et al., 2015; Marangos et al., 2015; Collins and Jones, 2016). The implication is that if the SAC is compromised, chromosomes may divide, despite DNA lesions. The challenge is to determine what endogenous lesions may trigger the SAC and whether such lesions would generate aneuploidy, as opposed to chromosome breakage and cellular arrest. If this is an important function of the SAC in the origin of human aneuploidies, then one would expect to see damaged chromosomes in oocytes.

In addition to non-canonical crosstalk between the DNA damage checkpoint and the SAC, the mitotic surveillance pathway blocks cell-cycle progression in response to prolonged mitosis (M-phase) and the loss of centrosomes (reviewed in Lambrus and Holland, 2017). Intriguingly, DNA damage response genes 53BP1 and USP28 act upstream of p53 to inhibit the proliferation of daughter cells that become aneuploid (Hinchcliffe et al., 2016). We do not know whether this pathway is active in meiosis, but it raises the interesting question of how centrosomes become ‘lost’ in human oocytes and how this is tolerated. One potential caveat is that mouse models targetting 53BP1 are viable, questioning the effect that the mitotic surveillance pathway has in vivo (Fernandez-Capetillo et al., 2002).

Do the cellular checkpoints that are active in murine oocytes contribute towards human aneuploidies? The oocyte-specific facets of cell-cycle control have been proposed to create vulnerability to segregation errors (Nagaoka et al., 2011). There is also evidence that the levels of several checkpoint proteins, such as Mad2, is lowered in aged mouse oocytes. This may lead to decreased error correction, when spindle organization is impaired during meiosis I (Yun et al., 2014). However, many genes show transcriptional dysregulation in aged human oocytes (Grondahl et al., 2010), and improved models will need to correlate several factors including phenotypic expression in haploinsufficiency models in mouse. Variants in both Aurora B and C were recently mapped and functional characterization of the variants, when overexpressed in mouse oocytes, suggested that the different alleles may support different SAC strength (Nguyen and Schindler, 2017; Nguyen et al., 2017). Collectively, such studies suggest that modulation to cell-cycle checkpoints may be important regulators of genome integrity and aneuploidy in the natural population.

Maturation arrest and spindle assembly defects

Although much focus has been on the SAC, recent observations suggest that the spindle itself may cause arrest and, potentially, segregation errors in meiosis. Feng and colleagues investigated the genetic causes of several families showing complete oocyte maturation arrest at the MI stage (Feng et al., 2016). The authors found seven different mutations in the β-tubulin-encoding gene, TUBB8, in women with primary infertility due to meiosis I arrest of their oocytes. The TUBB8 mutations were paternally inherited in five of the families whereas de novo mutations in TUBB8 occurred in the affected women of the other two families. In the same study, the authors not only found that TUBB8 is primarily expressed in human oocytes and embryos, they also determined that the TUBB8 protein is an essential and major component of the oocyte spindle. The introduction of mutant protein, but not wild type TUBB8, into HeLa cells and yeast cells caused altered microtubule dynamics. Mutant TUBB8 protein was also dominant when expressed in mouse and human oocytes, causing maturation arrest, thereby recapitulating the phenotype observed in the women presenting with primary infertility. The study has set a high bar for the functional confirmation that the mutations mapped to specific genes, in this case TUBB8, are the likely cause of pathogenicity.

Subsequent studies of mutations mapped in TUBB8 have revealed further complexities in the role of the meiosis-specific β-tubulin. The phenotypes include embryonic arrest (Chen et al., 2016; Feng et al., 2016). It is currently unknown whether the arrested preimplantation embryos are aneuploid and whether the mature oocytes that do complete the first division have a normal chromosome content. Given the severe spindle defects, it would not be unreasonable to speculate that the MII oocytes could be aneuploid. Assessing chromosome segregation when TUBB8 is perturbed may yield important insights into human aneuploidies.

Influence of genetic factors on human female meiosis

The J curve is a population average. The likelihood of producing an aneuploid oocyte cannot be predicted reliably by maternal age alone. There is a significant variation in aneuploidy rates amongst individuals of the same reproductive age (Franasiak et al., 2014a,b). Relatively young women (<35 years old) appear to have unexpectedly high levels of aneuploid oocytes compared to many women considered to be of AMA (>35 years old). The only genome-wide association study of chromosome errors in human preimplantation embryos identified a quantitative trait locus for mitotic errors, but did not find any quantitative trait loci that were associated with maternal aneuploidies, at least not any that were significantly elevated above the false discovery rate (McCoy et al., 2015). A target sequencing approach identified putative Aurora B and C variants, as discussed above (Nguyen et al., 2017). Given the large volume of data generated from the increased application of aneuploidy testing in ART cycles, further studies may provide insight into this important medical and societal issue in the near future.

Several observations suggest that genetic factors could predispose to both the general baseline and age-related elevation in aneuploidy conceptions. A recent genome-wide screen for new meiotic genes in mouse oocytes revealed hundreds of genes, whose depletion by RNAi affected chromosome segregation (Pfender et al., 2015). This suggests that conducting refined analyses in human oocytes and population-based studies may yet yield new molecular targets. Studies in mice suggest that heterozygosity of SMC1β, a conserved meiosis-specific cohesin subunit, predisposes to aneuploidy (Murdoch et al., 2013). Deletion of both copies of SMC1β predisposes to age-related loss of bivalent structures and therefore to aneuploidy in mouse oocytes (Hodges et al., 2005). The haploinsufficiency studies are important because they suggest dosage sensitivity. This is particularly relevant in human populations where complete deletions (homozygous) of gene activities are relatively rare and usually only found in consanguineous families (O’Driscoll, 2008).

There have been several studies attempting to investigate the link between ovarian aging (premature ovarian insufficiency) and chromosomal aging of human oocytes. Currently, there are no firm links between the two. However, we did find a putative SNP where women with mutations that truncate the product of SYPC3 were at risk of recurrent miscarriage due to aneuploidy in the foetus (Bolor et al., 2009). The findings were interesting because the function of SYCP3 function in murine meiosis is well defined, as it acts as a chromosomal scaf-fold protein that forms a part of the synaptonemal complex and promotes crossing over (Lammers et al., 1994). The association between recurrent miscarriage and mutation in SYCP3, however, was not reproduced by a second study of the same SNP in the same ethnic population (Mizutani et al., 2011) nor in a different population (Hanna et al., 2012).

In summary, there are currently several molecular models from mouse studies that firmly implicate multiple different mechanisms affecting chromosomes and spindles as well as lowered cell-cycle control. However, clinical associations between human aneuploidy and their prognostic value remain to be determined. Once defined, the multifactorial nature of aneuploidy may help the development of genetic biomarkers for increased susceptibility to aneuploidies in women of young reproductive age, so that they may opt for preventive measures, or it may even lead to cures.

Artificial gametes and chromosome therapy in living oocytes and preimplantation embryos

The high incidence of aneuploidy in human oocytes has given rise to extensive prenatal screening programmes as well as genetic testing to select euploid embryos for transfer in IVF clinics (reviewed in Vermeesch et al., 2016). Preimplantation genetic testing (Fig. 4) is only a diagnostic procedure that increases the efficiency of treatment but does not increase the pregnancy chance for a given stimulation cycle. This is important since attrition is still the major factor why couples do not obtain a pregnancy in IVF settings. However, aneuploidy testing does not change the genetic status of the oocytes that a woman produces. Once a woman reaches the point where she no longer produces any chromosomally normal oocytes, aneuploidy screening no longer provides any benefit in terms of pregnancy success. In many countries, this limits the age at which women may undergo an IVF treatment.

How do we improve reproductive success and reduce the effects of maternal aging in the natural population as well as in the clinic? Identification of lifestyle factors that affect natural conception is important. Several factors including smoking, irradiation, oral contraceptives and low socioeconomic status (Christianson et al., 2004; Hunter et al., 2013) have been implicated but their molecular basis has yet to be elucidated. Disentangling the factors that influence aneuploidy may provide us with lifestyle interventions to reduce miscarriage rates and may move the J curve to the right and prevent the early truncation of reproductive lifespan caused by aneuploidy.

One lifestyle intervention may be to target cellular responses that are activated in response to aging, obesity or high lipid diets. Several studies in mice have suggested that targeting the unfolded protein response, which is activated by stress in the endoplasmic reticulum (ER), improves maturation of oocytes as well as developmental outcomes of the offspring. When follicles from obese mice or obese females were exposed in vitro or in vivo, respectively, to salubrinal, this improved maturation rates, conceptions, and health of the offspring (Wu et al., 2010, 2012, 2015). In vivo, salubrinal was only supplied for a very short duration prior to ovulation (Wu et al., 2015), suggesting that anovulation in obese females can be reversed without major interventions. Given that ER stress is implicated in a wide range of cellular pathologies (Martinez et al., 2017), the strength of the in vivo and in vitro data from murine models warrants preclinical assessment of ER inhibitors in improving maturation efficiency of human oocytes.

Another avenue of research is to understand how much of our knowledge from aging models in mice we can translate to humans, and in particular what may be relevant for reproductive aging. There are several progeria models of mouse, including DDR genes (e.g. Murga et al., 2009), that may be interesting to assess given that common variants of several DDR genes were implicated in determining the age of onset of natural menopause (Day et al., 2015). Premature ovarian insufficiency has also been linked to mutations in the DDR genes, including BRCA1 (Titus et al., 2013) and meiosis-specific STAG3 whose protein (Caburet et al., 2014) is a component of meiotic cohesin complexes. Deletion of STAG3 causes persistent DNA damage and failure of chromosome synapsis in meiotic cells (Caburet et al., 2014; Fukuda et al., 2014; Hopkins et al., 2014; Winters et al., 2014), like many other cohesin mutants (McNicoll et al., 2013). However, it is unclear whether the DDR solely dictates maintenance of the follicle pool, or, whether this complex network of more than 450 genes (Pearl et al., 2015) also influences chromosome segregation and therefore, aneuploidy.

Chromosome therapy is a functional correction of aneuploidies in living cells. This has been achieved with two different molecular strategies. Jiang et al. exploited the natural mechanism of XIST, a non-conding RNA that drives the transcriptional silencing of one of the X-chromosomes in females by inducing heterochromatic modifications to the inactive X chromosome (Jiang et al., 2013). The authors transcribed XIST from the DYRK1A locus of chromosome 21 in iPS cells from a Down Syndrome individual. After induction of the ectopically inserted XIST, regions of the targeted chromosome 21 became heterochromatized and transcriptionally silenced, and a ‘chromosome 21 Barr body’ was detectable. The deficit in cell proliferation and differentiation of iPS cells into neural progenitor cells was also reversed by chromosome 21 repression. The heterochromatic silencing was stably maintained after the initial XIST expression induction and the system was compatible with natural female X inactivation, making this a promising strategy for translating dosage compensation to individuals with Down Syndrome.

Another strategy to achieve correct chromosome number is based on the use of a mammalian-specific gene, ZSCAN4 (zinc finger and SCAN domain containing 4) (Akiyama et al., 2015; Amano et al., 2015). We currently only have data from mouse, where it is expressed at the 2-cell stage of preimplantation embryos and is required for genome stability and maintenance of a normal karyotype in mouse ES cells. Using mRNA and Sendai virus vectors encoding human ZSCAN4, trisomy 18 or 21 could be corrected to euploid, apparently without affecting other chromosomes.

One intriguing possibility is whether such new biologics could be developed to enhance the efficiency of ZSCAN4 during oogenesis of GV oocytes in vitro and/or preimplantation development of human embryos.

Alternatives to chromosome therapy include mRNA or use of inhibitors during in vitro maturation or embryo development. Elucidation of putative mechanisms where factors may be rate-limiting for accurate chromosome segregation raises the possibility of supplying the protein or mRNA to the oocyte. Conversely, elevated levels of inhibitory proteins could be treated by use of inhibitors. Such procedures would not alter the genetic make-up of the oocyte, but would facilitate improved genetic integrity of human embryos generated in vitro. Even though, these are only mere perspectives at present, such developments, once available, would counteract the natural mechanism of reproductive aging and increase the cumulative live birth rate per started cycle.

Future Perspectives

Our increasing knowledge of human aneuploidies and their origins together with advances in technologies are allowing unprecedented insight into chromosome errors in human conceptions. The challenge is to translate this knowledge into improved diagnosis and therapies for precision medicine that allows women to make informed choices about their reproductive health. Improving the efficiency of medically assisted reproduction is critical and the future is promising with regards to developing interventions to improve the genetic quality of oocytes, especially as women age reproductively. Improving reproductive aging to match the increased lifespan currently experienced in the human population is important for fertility and health issues associated with the cessation of reproductive functions in women.

One concern that we still need to address is that many of our studies on human gametes are based on oocytes obtained from gonadotrophin-stimulated women. Although there are studies that suggest that gentle stimulation regimes do not affect aneuploidy rates (Plachot, 2001; Labarta et al., 2012), and therefore, presumably, segregation of chromosomes, other studies have found significant effects of gonadotrophin stimulation regime on aneuploidy (Baart et al., 2007; Rubio et al., 2010). It remains to be determined whether the new findings on meiotic segregation are restricted to IVF oocytes or if they are a universal phenomenon. Nevertheless, as our knowledge about meiosis increases in human oocytes from medically assisted reproduction, it is also possible that clinical material that is currently not used for treatment may become an option for fertility treatment, in case mature oocytes are scarce or stimulation is not possible. Basic research on female meiosis may therefore contribute towards further developments in the in vitro maturation treatment of immature oocytes (Coticchio et al., 2015).

Acknowledgements

We thank our colleagues, especially Dr Xavi Aran, for insightful comments and proof-reading of the manuscript. We apologize to our colleagues, whose work we could not cite due to limitations of space.

Authors’ roles

A.C. and E.R.H. conceived the study and wrote the manuscript. D.C. performed manuscript drafting and data preparation. F.M.U. provided critical discussion. L.R. participated in study design and provided critical discussion of the manuscript.

Funding

No specific funding was used for this study.

Conflict of interest

E.R.H. is funded by a Novo Nordisk Young Investigator Award. E.R.H. receives funding from Illumina Ltd. on which the MeioMapping is based. GENERA and GENETYX provide a PGT-A service to their patients. The remaining authors have no conflicts of interest.

References

Akiyama
T
,
Xin
L
,
Oda
M
,
Sharov
AA
,
Amano
M
,
Piao
Y
,
Cadet
JS
,
Dudekula
DB
,
Qian
Y
,
Wang
W
et al. .
Transient bursts of Zscan4 expression are accompanied by the rapid derepression of heterochromatin in mouse embryonic stem cells
.
DNA Res
2015
;
22
:
307
318
.

Amano
T
,
Jeffries
E
,
Amano
M
,
Ko
AC
,
Yu
H
,
Ko
MS
.
Correction of Down syndrome and Edwards syndrome aneuploidies in human cell cultures
.
DNA Res
2015
;
22
:
331
342
.

Andersen
CY
.
Success and challenges in fertility preservation after ovarian tissue grafting
.
Lancet
2015
;
385
:
1947
1948
.

Angell
RR
.
Predivision in human oocytes at meiosis I: a mechanism for trisomy formation in man
.
Hum Genet
1991
;
86
:
383
387
.

Angell
RR
.
Meiosis I in human oocytes
.
Cytogenet Genet
1995
;
69
:
266
272
.

Baart
EB
,
Martini
E
,
Eijkemans
MJ
,
Van Opstal
D
,
Beckers
NG
,
Verhoeff
A
,
Macklon
NS
,
Fauser
BC
.
Milder ovarian stimulation for in-vitro fertilization reduces aneuploidy in the human preimplantation embryo: a randomized controlled trial
.
Hum Reprod
2007
;
22
:
980
988
.

Baker
SM
,
Plug
AW
,
Prolla
TA
,
Bronner
CE
,
Harris
AC
,
Yao
X
,
Christie
DM
,
Monell
C
,
Arnheim
N
,
Bradley
A
et al. .
Involvement of mouse Mlh1 in DNA mismatch repair and meiotic crossing over
.
Nat Genet
1996
;
13
:
336
342
.

Battaglia
DE
,
Goodwin
P
,
Klein
NA
,
Soules
MR
.
Influence of maternal age on meiotic spindle assembly in oocytes from naturally cycling women
.
Hum Reprod
1996
;
11
:
2217
2222
.

Bolcun-Filas
E
,
Rinaldi
VD
,
White
ME
,
Schimenti
JC
.
Reversal of female infertility by Chk2 ablation reveals the oocyte DNA damage checkpoint pathway
.
Science
2014
;
343
:
533
536
.

Bolor
H
,
Mori
T
,
Nishiyama
S
,
Ito
Y
,
Hosoba
E
,
Inagaki
H
,
Kogo
H
,
Ohye
T
,
Tsutsumi
M
,
Kato
T
et al. .
Mutations of the SYCP3 gene in women with recurrent pregnancy loss
.
Am J Hum Genet
2009
;
84
:
14
20
.

Caburet
S
,
Arboleda
VA
,
Llano
E
,
Overbeek
PA
,
Barbero
JL
,
Oka
K
,
Harrison
W
,
Vaiman
D
,
Ben-Neriah
Z
,
Garcia-Tunon
I
et al. .
Mutant cohesin in premature ovarian failure
.
N Engl J Med
2014
;
370
:
943
949
.

Campbell
CL
,
Furlotte
NA
,
Eriksson
N
,
Hinds
D
,
Auton
A
.
Escape from crossover interference increases with maternal age
.
Nat Commun
2015
;
6
:
6260
.

Capalbo
A
,
Bono
S
,
Spizzichino
L
,
Biricik
A
,
Baldi
M
,
Colamaria
S
,
Ubaldi
FM
,
Rienzi
L
,
Fiorentino
F
.
Sequential comprehensive chromosome analysis on polar bodies, blastomeres and trophoblast: insights into female meiotic errors and chromosomal segregation in the preimplantation window of embryo development
.
Hum Reprod
2013
;
28
:
509
518
.

Capalbo
A
,
Rienzi
L
,
Cimadomo
D
,
Maggiulli
R
,
Elliott
T
,
Wright
G
,
Nagy
ZP
,
Ubaldi
FM
.
Correlation between standard blastocyst morphology, euploidy and implantation: an observational study in two centers involving 956 screened blastocysts
.
Hum Reprod
2014
;
9
:
1173
1181
.

Capalbo
A
,
Treff
NR
,
Cimadomo
D
,
Tao
X
,
Upham
K
,
Ubaldi
FM
,
Rienzi
L
,
Scott
RT
Jr
.
Comparison of array comparative genomic hybridization and quantitative real-time PCR-based aneuploidy screening of blastocyst biopsies
.
EJHG
2015
;
23
:
901
906
.

Capalbo
A
,
Ubaldi
FM
,
Cimadomo
D
,
Maggiulli
R
,
Patassini
C
,
Dusi
L
,
Sanges
F
,
Buffo
L
,
Venturella
R
,
Rienzi
L
.
Consistent and reproducible outcomes of blastocyst biopsy and aneuploidy screening across different biopsy practitioners: a multicentre study involving 2586 embryo biopsies
.
Hum Reprod
2016
;
31
:
199
208
.

Chen
B
,
Li
B
,
Li
D
,
Yan
Z
,
Mao
X
,
Xu
Y
,
Mu
J
,
Li
Q
,
Jin
L
,
He
L
et al. .
Novel mutations and structural deletions in TUBB8: expanding mutational and phenotypic spectrum of patients with arrest in oocyte maturation, fertilization or early embryonic development
.
Hum Reprod
2016
;
32
:
457
464
.

Chen
M
,
Wei
S
,
Hu
J
,
Quan
S
.
Can comprehensive chromosome screening technology improve IVF/ICSI Outcomes? A Meta-Analysis
.
PLoS One
2015
;
10
:
e0140779
.

Cheng
EY
,
Hunt
PA
,
Naluai-Cecchini
TA
,
Fligner
CL
,
Fujimoto
VY
,
Pasternack
TL
,
Schwartz
JM
,
Steinauer
JE
,
Woodruff
TJ
,
Cherry
SM
et al. .
Meiotic recombination in human oocytes
.
PLoS Genet
2009
;
5
:
e1000661
.

Chiang
T
,
Duncan
FE
,
Schindler
K
,
Schultz
RM
,
Lampson
MA
.
Evidence that weakened centromere cohesion is a leading cause of age-related aneuploidy in oocytes
.
Curr Biol
2010
;
20
:
1522
1528
.

Christianson
RE
,
Sherman
SL
,
Torfs
CP
.
Maternal meiosis II nondisjunction in trisomy 21 is associated with maternal low socioeconomic status
.
Genet Med
2004
;
6
:
487
494
.

Christopikou
D
,
Tsorva
E
,
Economou
K
,
Shelley
P
,
Davies
S
,
Mastrominas
M
,
Handyside
AH
.
Polar body analysis by array comparative genomic hybridization accurately predicts aneuploidies of maternal meiotic origin in cleavage stage embryos of women of advanced maternal age
.
Hum Reprod
2013
;
28
:
1426
1434
.

Cloutier
JM
,
Mahadevaiah
SK
,
ElInati
E
,
Toth
A
,
Turner
J
.
Mammalian meiotic silencing exhibits sexually dimorphic features
.
Chromosoma
2015
;
125
:
215
226
.

Collins
JK
,
Jones
KT
.
DNA damage responses in mammalian oocytes
.
Reproduction
2016
;
152
:
R15
R22
.

Collins
JK
,
Lane
SI
,
Merriman
JA
,
Jones
KT
.
DNA damage induces a meiotic arrest in mouse oocytes mediated by the spindle assembly checkpoint
.
Nat Commun
2015
;
6
:
8553
.

Coop
G
,
Wen
X
,
Ober
C
,
Pritchard
JK
,
Przeworski
M
.
High-resolution mapping of crossovers reveals extensive variation in fine-scale recombination patterns among humans
.
Science
2008
;
319
:
1395
1398
.

Coticchio
G
,
Dal Canto
M
,
Mignini Renzini
M
,
Guglielmo
MC
,
Brambillasca
F
,
Turchi
D
,
Novara
PV
,
Fadini
R
.
Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization
.
Hum Reprod Update
2015
;
21
:
427
454
.

Day
FR
,
Ruth
KS
,
Thompson
DJ
,
Lunetta
KL
,
Pervjakova
N
,
Chasman
DI
,
Stolk
L
,
Finucane
HK
,
Sulem
P
,
Bulik-Sullivan
B
et al. .
Large-scale genomic analyses link reproductive aging to hypothalamic signaling, breast cancer susceptibility and BRCA1-mediated DNA repair
.
Nat Genet
2015
;
47
:
1294
1303
.

Duncan
FE
,
Hornick
JE
,
Lampson
MA
,
Schultz
RM
,
Shea
LD
,
Woodruff
TK
.
Chromosome cohesion decreases in human eggs with advanced maternal age
.
Aging Cell
2012
;
11
:
1121
1124
.

Edelmann
W
,
Cohen
PE
,
Kane
M
,
Lau
K
,
Morrow
B
,
Bennett
S
,
Umar
A
,
Kunkel
T
,
Cattoretti
G
,
Chaganti
R
et al. .
Meiotic pachytene arrest in MLH1-deficient mice
.
Cell
1996
;
85
:
1125
1134
.

Erickson
JD
.
Down syndrome, paternal age, maternal age and birth order
.
Ann Hum Genet
1978
;
41
:
289
298
.

Feng
R
,
Sang
Q
,
Kuang
Y
,
Sun
X
,
Yan
Z
,
Zhang
S
,
Shi
J
,
Tian
G
,
Luchniak
A
,
Fukuda
Y
et al. .
Mutations in TUBB8 and Human Oocyte Meiotic Arrest
.
N Engl J Med
2016
;
374
:
223
232
.

Fernandez-Capetillo
O
,
Chen
HT
,
Celeste
A
,
Ward
I
,
Romanienko
PJ
,
Morales
JC
,
Naka
K
,
Xia
Z
,
Camerini-Otero
RD
,
Motoyama
N
et al. .
DNA damage-induced G2-M checkpoint activation by histone H2AX and 53BP1
.
Nat Cell Biol
2002
;
4
:
993
997
.

Ford
CE
,
Jones
KW
,
Polani
PE
,
De Almeida
JC
,
Briggs
JH
.
A sex-chromosome anomaly in a case of gonadal dysgenesis (Turner's syndrome)
.
Lancet
1959
a;
1
:
711
713
.

Ford
CE
,
Polani
PE
,
Briggs
JH
,
Bishop
PM
.
A presumptive human XXY/XX mosaic
.
Nature
1959
b;
183
:
1030
1032
.

Forman
EJ
,
Treff
NR
,
Stevens
JM
,
Garnsey
HM
,
Katz-Jaffe
MG
,
Scott
RT
Jr
,
Schoolcraft
WB
.
Embryos whose polar bodies contain isolated reciprocal chromosome aneuploidy are almost always euploid
.
Hum Reprod
2013
;
28
:
502
508
.

Fragouli
E
,
Escalona
A
,
Gutierrez-Mateo
C
,
Tormasi
S
,
Alfarawati
S
,
Sepulveda
S
,
Noriega
L
,
Garcia
J
,
Wells
D
,
Munne
S
.
Comparative genomic hybridization of oocytes and first polar bodies from young donors
.
Reprod Med Online
2009
;
19
:
228
237
.

Franasiak
JM
,
Forman
EJ
,
Hong
KH
,
Werner
MD
,
Upham
KM
,
Treff
NR
,
Scott
RT
.
Aneuploidy across individual chromosomes at the embryonic level in trophectoderm biopsies: changes with patient age and chromosome structure
.
J Ass Reprod Genet
2014
a;
31
:
1501
1509
.

Franasiak
JM
,
Forman
EJ
,
Hong
KH
,
Werner
MD
,
Upham
KM
,
Treff
NR
,
Scott
RT
Jr
.
The nature of aneuploidy with increasing age of the female partner: a review of 15,169 consecutive trophectoderm biopsies evaluated with comprehensive chromosomal screening
.
Fertil Steril
2014
b;
101
:
656
663 e651
.

Fukuda
T
,
Fukuda
N
,
Agostinho
A
,
Hernandez-Hernandez
A
,
Kouznetsova
A
,
Hoog
C
.
STAG3-mediated stabilization of REC8 cohesin complexes promotes chromosome synapsis during meiosis
.
EMBO J
2014
;
33
:
1243
1255
.

Gabriel
AS
,
Hassold
TJ
,
Thornhill
AR
,
Affara
NA
,
Handyside
AH
,
Griffin
DK
.
An algorithm for determining the origin of trisomy and the positions of chiasmata from SNP genotype data
.
Chromosome Res
2011
;
19
:
155
163
.

Garcia-Cruz
R
,
Brieno
MA
,
Roig
I
,
Grossmann
M
,
Velilla
E
,
Pujol
A
,
Cabero
L
,
Pessarrodona
A
,
Barbero
JL
,
Garcia Caldes
M
.
Dynamics of cohesin proteins REC8, STAG3, SMC1 beta and SMC3 are consistent with a role in sister chromatid cohesion during meiosis in human oocytes
.
Hum Reprod
2010
a;
25
:
2316
2327
.

Garcia-Cruz
R
,
Casanovas
A
,
Brieno-Enriquez
M
,
Robles
P
,
Roig
I
,
Pujol
A
,
Cabero
L
,
Durban
M
,
Garcia Caldes
M
.
Cytogenetic analyses of human oocytes provide new data on non-disjunction mechanisms and the origin of trisomy 16
.
Hum Reprod
2010
b;
25
:
179
191
.

Grondahl
ML
,
Yding Andersen
C
,
Bogstad
J
,
Nielsen
FC
,
Meinertz
H
,
Borup
R
.
Gene expression profiles of single human mature oocytes in relation to age
.
Hum Reprod
2010
;
25
:
957
968
.

Hached
K
,
Xie
SZ
,
Buffin
E
,
Cladiere
D
,
Rachez
C
,
Sacras
M
,
Sorger
PK
,
Wassmann
K
.
Mps1 at kinetochores is essential for female mouse meiosis I
.
Development
2011
;
138
:
2261
2271
.

Handyside
AH
,
Montag
M
,
Magli
MC
,
Repping
S
,
Harper
J
,
Schmutzler
A
,
Vesela
K
,
Gianaroli
L
,
Geraedts
J
.
Multiple meiotic errors caused by predivision of chromatids in women of advanced maternal age undergoing in vitro fertilisation
.
EJHG
2012
;
20
:
742
747
.

Hanna
CW
,
Blair
JD
,
Stephenson
MD
,
Robinson
WP
.
Absence of SYCP3 mutations in women with recurrent miscarriage with at least one trisomic miscarriage
.
Reprod Med Online
2012
;
24
:
251
253
.

Hassold
T
,
Chen
N
,
Funkhouser
J
,
Jooss
T
,
Manuel
B
,
Matsuura
J
,
Matsuyama
A
,
Wilson
C
,
Yamane
JA
,
Jacobs
PA
.
A cytogenetic study of 1000 spontaneous abortions
.
Ann Hum Genet
1980
;
44
:
151
178
.

Hassold
T
,
Hunt
P
.
To err (meiotically) is human: the genesis of human aneuploidy
.
Nat Rev Genet
2001
;
2
:
280
291
.

Henderson
SA
,
Edwards
RG
.
Chiasma frequency and maternal age in mammals
.
Nature
1968
;
218
:
22
28
.

Hinchcliffe
EH
,
Day
CA
,
Karanjeet
KB
,
Fadness
S
,
Langfald
A
,
Vaughan
KT
,
Dong
Z
.
Chromosome missegregation during anaphase triggers p53 cell cycle arrest through histone H3.3 Ser31 phosphorylation
.
Nat Cell Biol
2016
;
18
:
668
675
.

Hodges
CA
,
Revenkova
E
,
Jessberger
R
,
Hassold
TJ
,
Hunt
PA
.
SMC1beta-deficient female mice provide evidence that cohesins are a missing link in age-related nondisjunction
.
Nat Genet
2005
;
37
:
1351
1355
.

Holubcova
Z
,
Blayney
M
,
Elder
K
,
Schuh
M
.
Human oocytes. Error-prone chromosome-mediated spindle assembly favors chromosome segregation defects in human oocytes
.
Science
2015
;
348
:
1143
1147
.

Homer
HA
,
McDougall
A
,
Levasseur
M
,
Murdoch
AP
,
Herbert
M
.
Mad2 is required for inhibiting securin and cyclin B degradation following spindle depolymerisation in meiosis I mouse oocytes
.
Reproduction
2005
;
130
:
829
843
.

Hopkins
J
,
Hwang
G
,
Jacob
J
,
Sapp
N
,
Bedigian
R
,
Oka
K
,
Overbeek
P
,
Murray
S
,
Jordan
PW
.
Meiosis-specific cohesin component, Stag3 is essential for maintaining centromere chromatid cohesion, and required for DNA repair and synapsis between homologous chromosomes
.
PLoS Genet
2014
;
10
:
e1004413
.

Hou
Y
,
Fan
W
,
Yan
L
,
Li
R
,
Lian
Y
,
Huang
J
,
Li
J
,
Xu
L
,
Tang
F
,
Xie
XS
et al. .
Genome analyses of single human oocytes
.
Cell
2013
;
155
:
1492
1506
.

Hunt
PA
,
Hassold
TJ
.
Human female meiosis: what makes a good egg go bad?
Trends Genet
2008
;
24
:
86
93
.

Hunter
JE
,
Allen
EG
,
Shin
M
,
Bean
LJ
,
Correa
A
,
Druschel
C
,
Hobbs
CA
,
O’Leary
LA
,
Romitti
PA
,
Royle
MH
et al. .
The association of low socioeconomic status and the risk of having a child with Down syndrome: a report from the National Down Syndrome Project
.
Genet Med
2013
;
15
:
698
705
.

Hunter
N
,
Borts
RH
.
Mlh1 is unique among mismatch repair proteins in its ability to promote crossing-over during meiosis
.
Genes Dev
1997
;
11
:
1573
1582
.

Hwang
G
,
Sun
F
,
O’Brien
M
,
Eppig
JJ
,
Handel
MA
,
Jordan
PW
.
SMC5/6 is required for the formation of segregation-competent bivalent chromosomes during meiosis I in mouse oocytes
.
Development
2017
;
144
:
1648
1660
.

Jackson
SP
,
Bartek
J
.
The DNA-damage response in human biology and disease
.
Nature
2009
;
461
:
1071
1078
.

Jacobs
PA
,
Baikie
AG
,
Court Brown
WM
,
Strong
JA
.
The somatic chromosomes in mongolism
.
Lancet
1959
;
1
:
710
.

Jacobs
PA
,
Strong
JA
.
A case of human intersexuality having a possible XXY sex-determining mechanism
.
Nature
1959
;
183
:
302
303
.

Jagiello
GM
,
Ducayen
M
,
Fang
JS
,
Graffeo
J
. Cytogenetic observations in mammalian oocytes. In:
Pearson
PL
,
Lewis
KR
(eds).
Chrom Today V
.
New York, NY
:
Wiley
,
1975
;
43
63
.

Jessberger
R
.
Age-related aneuploidy through cohesion exhaustion
.
EMBO Rep
2012
;
13
:
539
546
.

Jiang
J
,
Jing
Y
,
Cost
GJ
,
Chiang
JC
,
Kolpa
HJ
,
Cotton
AM
,
Carone
DM
,
Carone
BR
,
Shivak
DA
,
Guschin
DY
et al. .
Translating dosage compensation to trisomy 21
.
Nature
2013
;
500
:
296
300
.

Keeney
S
,
Lange
J
,
Mohibullah
N
.
Self-organization of meiotic recombination initiation: general principles and molecular pathways
.
Ann Rev Genet
2014
;
48
:
187
214
.

Kim
J
,
Ishiguro
K
,
Nambu
A
,
Akiyoshi
B
,
Yokobayashi
S
,
Kagami
A
,
Ishiguro
T
,
Pendas
AM
,
Takeda
N
,
Sakakibara
Y
et al. .
Meikin is a conserved regulator of meiosis-I-specific kinetochore function
.
Nature
2015
;
517
:
466
471
.

Koehler
KE
,
Boulton
CL
,
Collins
HE
,
French
RL
,
Herman
KC
,
Lacefield
SM
,
Madden
LD
,
Schuetz
CD
,
Hawley
RS
.
Spontaneous X chromosome MI and MII nondisjunction events in Drosophila melanogaster oocytes have different recombinational histories
.
Nat Genet
1996
;
14
:
406
414
.

Koehler
KE
,
Schrump
SE
,
Cherry
JP
,
Hassold
TJ
,
Hunt
PA
.
Near-human aneuploidy levels in female mice with homeologous chromosomes
.
Curr Biol
2006
;
16
:
R579
R580
.

Kong
A
,
Barnard
J
,
Gudbjartsson
DF
,
Thorleifsson
G
,
Jonsdottir
G
,
Sigurdardottir
S
,
Richardsson
B
,
Jonsdottir
J
,
Thorgeirsson
T
,
Frigge
ML
et al. .
Recombination rate and reproductive success in humans
.
Nat Genet
2004
;
36
:
1203
1206
.

Kouznetsova
A
,
Lister
L
,
Nordenskjold
M
,
Herbert
M
,
Hoog
C
.
Bi-orientation of achiasmatic chromosomes in meiosis I oocytes contributes to aneuploidy in mice
.
Nat Genet
2007
;
39
:
966
968
.

Labarta
E
,
Bosch
E
,
Alama
P
,
Rubio
C
,
Rodrigo
L
,
Pellicer
A
.
Moderate ovarian stimulation does not increase the incidence of human embryo chromosomal abnormalities in in vitro fertilization cycles
.
J Clin Endocrinol Metab
2012
;
97
:
E1987
E1994
.

Lagirand-Cantaloube
J
,
Ciabrini
C
,
Charrasse
S
,
Ferrieres
A
,
Castro
A
,
Anahory
T
,
Lorca
T
.
Loss of centromere cohesion in aneuploid human oocytes correlates with decreased kinetochore localization of the Sac Proteins Bub1 and Bubr1
.
Sci Rep
2017
;
7
:
44001
.

Lamb
NE
,
Sherman
SL
,
Hassold
TJ
.
Effect of meiotic recombination on the production of aneuploid gametes in humans
.
Cytogenet Genome Res
2005
;
111
:
250
255
.

Lambrus
BG
,
Holland
AJ
.
A New Mode of Mitotic Surveillance
.
Trends Cell Biol
2017
;
27
:
314
321
.

Lammers
JH
,
Offenberg
HH
,
van Aalderen
M
,
Vink
AC
,
Dietrich
AJ
,
Heyting
C
.
The gene encoding a major component of the lateral elements of synaptonemal complexes of the rat is related to X-linked lymphocyte-regulated genes
.
Mol Cell Biol
1994
;
14
:
1137
1146
.

Lee
J
,
Kitajima
TS
,
Tanno
Y
,
Yoshida
K
,
Morita
T
,
Miyano
T
,
Miyake
M
,
Watanabe
Y
.
Unified mode of centromeric protection by shugoshin in mammalian oocytes and somatic cells
.
Nat Cell Biol
2008
;
10
:
42
52
.

Lejeune
J
,
Gautier
M
,
Turpin
R
.
[Study of somatic chromosomes from 9 mongoloid children]
.
C R Hebd Seances Acad Sci
1959
;
248
:
1721
1722
.

LeMaire-Adkins
R
,
Hunt
PA
.
Nonrandom segregation of the mouse univalent X chromosome: evidence of spindle-mediated meiotic drive
.
Genetics
2000
;
156
:
775
783
.

Lenzi
ML
,
Smith
J
,
Snowden
T
,
Kim
M
,
Fishel
R
,
Poulos
BK
,
Cohen
PE
.
Extreme heterogeneity in the molecular events leading to the establishment of chiasmata during meiosis i in human oocytes
.
Am J Hum Genet
2005
;
76
:
112
127
.

Lister
LM
,
Kouznetsova
A
,
Hyslop
LA
,
Kalleas
D
,
Pace
SL
,
Barel
JC
,
Nathan
A
,
Floros
V
,
Adelfalk
C
,
Watanabe
Y
et al. .
Age-related meiotic segregation errors in mammalian oocytes are preceded by depletion of cohesin and Sgo2
.
Curr Biol
2010
;
20
:
1511
1521
.

Liu
L
,
Blasco
MA
,
Keefe
DL
.
Requirement of functional telomeres for metaphase chromosome alignments and integrity of meiotic spindles
.
EMBO Rep
2002
;
3
:
230
234
.

Lu
S
,
Zong
C
,
Fan
W
,
Yang
M
,
Li
J
,
Chapman
AR
,
Zhu
P
,
Hu
X
,
Xu
L
,
Yan
L
et al. .
Probing meiotic recombination and aneuploidy of single sperm cells by whole-genome sequencing
.
Science
2012
;
338
:
1627
1630
.

Mania
A
,
Mantzouratou
A
,
Delhanty
JD
,
Baio
G
,
Serhal
P
,
Sengupta
SB
.
Telomere length in human blastocysts
.
Reprod Med Online
2014
;
28
:
624
637
.

Marangos
P
,
Stevense
M
,
Niaka
K
,
Lagoudaki
M
,
Nabti
I
,
Jessberger
R
,
Carroll
J
.
DNA damage-induced metaphase I arrest is mediated by the spindle assembly checkpoint and maternal age
.
Nat Commun
2015
;
6
:
8706
.

Martin
RH
,
Mahadevan
MM
,
Taylor
PJ
,
Hildebrand
K
,
Long-Simpson
L
,
Peterson
D
,
Yamamoto
J
,
Fleetham
J
.
Chromosomal analysis of unfertilized human oocytes
.
J Reprod Fertil
1986
;
78
:
673
678
.

Martinez
G
,
Duran-Aniotz
C
,
Cabral-Miranda
F
,
Vivar
JP
,
Hetz
C
.
Endoplasmic reticulum proteostasis impairment in aging
.
Aging Cell
2017
;
16
:
615
623
.

McCoy
RC
,
Demko
Z
,
Ryan
A
,
Banjevic
M
,
Hill
M
,
Sigurjonsson
S
,
Rabinowitz
M
,
Fraser
HB
,
Petrov
DA
.
Common variants spanning PLK4 are associated with mitotic-origin aneuploidy in human embryos
.
Science
2015
;
348
:
235
238
.

McGuinness
BE
,
Anger
M
,
Kouznetsova
A
,
Gil-Bernabe
AM
,
Helmhart
W
,
Kudo
NR
,
Wuensche
A
,
Taylor
S
,
Hoog
C
,
Novak
B
et al. .
Regulation of APC/C activity in oocytes by a Bub1-dependent spindle assembly checkpoint
.
Curr Biol
2009
;
19
:
369
380
.

McNicoll
F
,
Stevense
M
,
Jessberger
R
.
Cohesin in gametogenesis
.
Curr Top Dev Biol
2013
;
102
:
1
34
.

Merriman
JA
,
Lane
SI
,
Holt
JE
,
Jennings
PC
,
Garcia-Higuera
I
,
Moreno
S
,
McLaughlin
EA
,
Jones
KT
.
Reduced chromosome cohesion measured by interkinetochore distance is associated with aneuploidy even in oocytes from young mice
.
Biol Reprod
2013
;
88
:
31
.

Middlebrooks
CD
,
Mukhopadhyay
N
,
Tinker
SW
,
Allen
EG
,
Bean
LJ
,
Begum
F
,
Chowdhury
R
,
Cheung
V
,
Doheny
K
,
Adams
M
et al. .
Evidence for dysregulation of genome-wide recombination in oocytes with nondisjoined chromosomes 21
.
Hum Mol Genet
2013
;
23
:
408
417
.

Mizutani
E
,
Suzumori
N
,
Ozaki
Y
,
Oseto
K
,
Yamada-Namikawa
C
,
Nakanishi
M
,
Sugiura-Ogasawara
M
.
SYCP3 mutation may not be associated with recurrent miscarriage caused by aneuploidy
.
Hum Reprod
2011
;
26
:
1259
1266
.

Murdoch
B
,
Owen
N
,
Stevense
M
,
Smith
H
,
Nagaoka
S
,
Hassold
T
,
McKay
M
,
Xu
H
,
Fu
J
,
Revenkova
E
et al. .
Altered cohesin gene dosage affects Mammalian meiotic chromosome structure and behavior
.
PLoS Genet
2013
;
9
:
e1003241
.

Murga
M
,
Bunting
S
,
Montana
MF
,
Soria
R
,
Mulero
F
,
Canamero
M
,
Lee
Y
,
McKinnon
PJ
,
Nussenzweig
A
,
Fernandez-Capetillo
O
.
A mouse model of ATR-Seckel shows embryonic replicative stress and accelerated aging
.
Nat Genet
2009
;
41
:
891
898
.

Nagaoka
SI
,
Hassold
TJ
,
Hunt
PA
.
Human aneuploidy: mechanisms and new insights into an age-old problem
.
Nat Rev Genet
2012
;
13
:
493
504
.

Nagaoka
SI
,
Hodges
CA
,
Albertini
DF
,
Hunt
PA
.
Oocyte-specific differences in cell-cycle control create an innate susceptibility to meiotic errors
.
Curr Biol
2011
;
21
:
651
657
.

Nguyen
AL
,
Marin
D
,
Zhou
A
,
Gentilello
AS
,
Smoak
EM
,
Cao
Z
,
Fedick
A
,
Wang
Y
,
Taylor
D
,
Scott
RT
et al. .
Identification and characterization of Aurora Kinase B and C variants associated with maternal aneuploidy
.
Mol Hum Reprod
2017
;
23
:
406
416
.

Nguyen
AL
,
Schindler
K
.
Specialize and divide (Twice): functions of three aurora kinase homologs in mammalian oocyte meiotic maturation
.
Trends Genet
2017
;
33
:
349
363
.

Niault
T
,
Hached
K
,
Sotillo
R
,
Sorger
PK
,
Maro
B
,
Benezra
R
,
Wassmann
K
.
Changing Mad2 levels affects chromosome segregation and spindle assembly checkpoint control in female mouse meiosis I
.
PLoS One
2007
;
2
:
e1165
.

Nicklas
RB
,
Koch
CA
.
Chromosome micromanipulation. 3. Spindle fiber tension and the reorientation of mal-oriented chromosomes
.
J Cell Biol
1969
;
43
:
40
50
.

Nishiyama
T
,
Ladurner
R
,
Schmitz
J
,
Kreidl
E
,
Schleiffer
A
,
Bhaskara
V
,
Bando
M
,
Shirahige
K
,
Hyman
AA
,
Mechtler
K
et al. .
Sororin mediates sister chromatid cohesion by antagonizing Wapl
.
Cell
2010
;
143
:
737
749
.

O’Driscoll
M
.
Haploinsufficiency of DNA damage response genes and their potential influence in human genomic disorders
.
Curr Genomics
2008
;
9
:
137
146
.

Ottolini
CS
,
Newnham
LJ
,
Capalbo
A
,
Natesan
SA
,
Joshi
HA
,
Cimadomo
D
,
Griffin
DK
,
Sage
K
,
Summers
MC
,
Thornhill
AR
et al. .
Genome-wide maps of recombination and chromosome segregation in human oocytes and embryos show selection for maternal recombination rates
.
Nat Genet
2015
;
47
:
727
735
.

Patel
J
,
Tan
SL
,
Hartshorne
GM
,
McAinsh
AD
.
Unique geometry of sister kinetochores in human oocytes during meiosis I may explain maternal age-associated increases in chromosomal abnormalities
.
Biol Open
2015
;
5
:
178
184
.

Pearl
LH
,
Schierz
AC
,
Ward
SE
,
Al-Lazikani
B
,
Pearl
FM
.
Therapeutic opportunities within the DNA damage response
.
Nat Rev Cancer
2015
;
15
:
166
180
.

Pellestor
F
,
Anahory
T
,
Hamamah
S
.
Effect of maternal age on the frequency of cytogenetic abnormalities in human oocytes
.
Cytogenet Genome Res
2005
;
111
:
206
212
.

Pellestor
F
,
Andreo
B
,
Arnal
F
,
Humeau
C
,
Demaille
J
.
Mechanisms of non-disjunction in human female meiosis: the co-existence of two modes of malsegregation evidenced by the karyotyping of 1397 in-vitro unfertilized oocytes
.
Hum Reprod
2002
;
17
:
2134
2145
.

Petronczki
M
,
Siomos
MF
,
Nasmyth
K
.
Un menage a quatre: the molecular biology of chromosome segregation in meiosis
.
Cell
2003
;
112
:
423
440
.

Pfender
S
,
Kuznetsov
V
,
Pasternak
M
,
Tischer
T
,
Santhanam
B
,
Schuh
M
.
Live imaging RNAi screen reveals genes essential for meiosis in mammalian oocytes
.
Nature
2015
;
524
:
239
242
.

Plachot
M
.
Chromosomal abnormalities in oocytes
.
Mol Cell Endocrinol
2001
;
183
:
S59
S63
.

Polani
PE
,
Jagiello
GM
.
Chiasmata, meiotic univalents, and age in relation to aneuploid imbalance in mice
.
Cytogenet Cell Genet
1976
;
16
:
505
529
.

Revenkova
E
,
Eijpe
M
,
Heyting
C
,
Hodges
CA
,
Hunt
PA
,
Liebe
B
,
Scherthan
H
,
Jessberger
R
.
Cohesin SMC1 beta is required for meiotic chromosome dynamics, sister chromatid cohesion and DNA recombination
.
Nat Cell BIol
2004
;
6
:
555
562
.

Rockmill
B
,
Voelkel-Meiman
K
,
Roeder
GS
.
Centromere-proximal crossovers are associated with precocious separation of sister chromatids during meiosis in Saccharomyces cerevisiae
.
Genetics
2006
;
174
:
1745
1754
.

Rowsey
R
,
Gruhn
J
,
Broman
KW
,
Hunt
PA
,
Hassold
T
.
Examining variation in recombination levels in the human female: a test of the production-line hypothesis
.
Am J Hum Genet
2014
;
95
:
108
112
.

Rubio
C
,
Bellver
J
,
Rodrigo
L
,
Castillón
G
,
Guillén
A
,
Vidal
C
,
Giles
J
,
Ferrando
M
,
Cabanillas
S
,
Remohí
J
,
Pellicer
A
,
Simón
C
.
In vitro fertilization with preimplantation genetic diagnosis for aneuploidies in advanced maternal age: a randomized, controlled study
.
Fertil Steril
2017
;
107
:
1122
1129
.

Rubio
C
,
Mercader
A
,
Alama
P
,
Lizan
C
,
Rodrigo
L
,
Labarta
E
,
Melo
M
,
Pellicer
A
,
Remohi
J
.
Prospective cohort study in high responder oocyte donors using two hormonal stimulation protocols: impact on embryo aneuploidy and development
.
Hum Reprod
2010
;
25
:
2290
2297
.

Sakakibara
Y
,
Hashimoto
S
,
Nakaoka
Y
,
Kouznetsova
A
,
Hoog
C
,
Kitajima
TS
.
Bivalent separation into univalents precedes age-related meiosis I errors in oocytes
.
Nat Commum
2015
;
6
:
7550
.

Salvaggio
CN
,
Forman
EJ
,
Garnsey
HM
,
Treff
NR
,
Scott
RT
Jr
.
Polar body based aneuploidy screening is poorly predictive of embryo ploidy and reproductive potential
.
J Ass Reprod Genet
2014
;
31
:
1221
1226
.

Scott
RT
Jr,
Ferry
K
,
Su
J
,
Tao
X
,
Scott
K
,
Treff
NR
.
Comprehensive chromosome screening is highly predictive of the reproductive potential of human embryos: a prospective, blinded, nonselection study
.
Fertil Steril
2012
;
97
:
870
875
.

Scott
RT
Jr,
Upham
KM
,
Forman
EJ
,
Zhao
T
,
Treff
NR
.
Cleavage-stage biopsy significantly impairs human embryonic implantation potential while blastocyst biopsy does not: a randomized and paired clinical trial
.
Fertil Steril
2013
;
100
:
624
630
.

Shibuya
H
,
Watanabe
Y
.
The meiosis-specific modification of mammalian telomeres
.
Cell Cycle
2014
;
13
:
2024
2028
.

Tease
C
,
Hartshorne
GM
,
Hulten
MA
.
Patterns of meiotic recombination in human fetal oocytes
.
Am J Hum Genet
2002
;
70
:
1469
1479
.

Titus
S
,
Li
F
,
Stobezki
R
,
Akula
K
,
Unsal
E
,
Jeong
K
,
Dickler
M
,
Robson
M
,
Moy
F
,
Goswami
S
et al. .
Impairment of BRCA1-related DNA double-strand break repair leads to ovarian aging in mice and humans
.
Sci Transl Med
2013
;
5
:
172ra121
.

Touati
SA
,
Buffin
E
,
Cladiere
D
,
Hached
K
,
Rachez
C
,
van Deursen
JM
,
Wassmann
K
.
Mouse oocytes depend on BubR1 for proper chromosome segregation but not for prophase I arrest
.
Nat Commun
2015
;
6
:
6946
.

Treff
NR
,
Su
J
,
Taylor
D
,
Scott
RT
Jr
.
Telomere DNA deficiency is associated with development of human embryonic aneuploidy
.
PLoS Genet
2011
;
7
:
e1002161
.

Tsutsumi
M
,
Fujiwara
R
,
Nishizawa
H
,
Ito
M
,
Kogo
H
,
Inagaki
H
,
Ohye
T
,
Kato
T
,
Fujii
T
,
Kurahashi
H
.
Age-related decrease of meiotic cohesins in human oocytes
.
PLoS One
2014
;
9
:
e96710
.

Turner
JM
.
Meiotic Silencing in Mammals
.
Ann Rev Genet
2015
;
49
:
395
412
.

Turner
S
,
Hartshorne
GM
.
Telomere lengths in human pronuclei, oocytes and spermatozoa
.
Mol Hum Reprod
2013
;
19
:
510
518
.

Turner
S
,
Wong
HP
,
Rai
J
,
Hartshorne
GM
.
Telomere lengths in human oocytes, cleavage stage embryos and blastocysts
.
Mol Hum Reprod
2010
;
16
:
685
694
.

van den Berg
IM
,
Eleveld
C
,
van der Hoeven
M
,
Birnie
E
,
Steegers
EA
,
Galjaard
RJ
,
Laven
JS
,
van Doorninck
JH
.
Defective deacetylation of histone 4 K12 in human oocytes is associated with advanced maternal age and chromosome misalignment
.
Hum Reprod
2011
;
26
:
1181
1190
.

van Echten-Arends
J
,
Mastenbroek
S
,
Sikkema-Raddatz
B
,
Korevaar
JC
,
Heineman
MJ
,
van der Veen
F
,
Repping
S
.
Chromosomal mosaicism in human preimplantation embryos: a systematic review
.
Hum Reprod Update
2011
;
17
:
620
627
.

Verlinsky
Y
,
Cieslak
J
,
Ivakhnenko
V
,
Evsikov
S
,
Wolf
G
,
White
M
,
Lifchez
A
,
Kaplan
B
,
Moise
J
,
Valle
J
et al. .
Chromosomal abnormalities in the first and second polar body
.
Mol Cell Endocrinol
2001
;
183
:
S47
S49
.

Vermeesch
JR
,
Voet
T
,
Devriendt
K
.
Prenatal and pre-implantation genetic diagnosis
.
Nat Rev Genet
2016
;
17
:
643
656
.

Wang
J
,
Fan
HC
,
Behr
B
,
Quake
SR
.
Genome-wide single-cell analysis of recombination activity and de novo mutation rates in human sperm
.
Cell
2012
;
150
:
402
412
.

Wang
S
,
Hassold
T
,
Hunt
P
,
White
MA
,
Zickler
D
,
Kleckner
N
,
Zhang
L
.
Inefficient Crossover Maturation Underlies Elevated Aneuploidy in Human Female Meiosis
.
Cell
2017
;
168
:
977
989 e917
.

Wassmann
K
,
Niault
T
,
Maro
B
.
Metaphase I arrest upon activation of the Mad2-dependent spindle checkpoint in mouse oocytes
.
Curr Biol
2003
;
13
:
1596
1608
.

Webster
A
,
Schuh
M
.
Mechanisms of Aneuploidy in Human Eggs
.
Trends Cell Biol
2016
;
27
:
55
68
.

Wells
D
,
Escudero
T
,
Levy
B
,
Hirschhorn
K
,
Delhanty
JD
,
Munne
S
.
First clinical application of comparative genomic hybridization and polar body testing for preimplantation genetic diagnosis of aneuploidy
.
Fertil Steril
2002
;
78
:
543
549
.

Winters
T
,
McNicoll
F
,
Jessberger
R
.
Meiotic cohesin STAG3 is required for chromosome axis formation and sister chromatid cohesion
.
EMBO J
2014
;
33
:
1256
1270
.

Wu
G
,
Hao
L
,
Han
Z
,
Gao
S
,
Latham
KE
,
de Villena
FP
,
Sapienza
C
.
Maternal transmission ratio distortion at the mouse Om locus results from meiotic drive at the second meiotic division
.
Genetics
2005
;
170
:
327
334
.

Wu
LL
,
Dunning
KR
,
Yang
X
,
Russell
DL
,
Lane
M
,
Norman
RJ
,
Robker
RL
.
High-fat diet causes lipotoxicity responses in cumulus-oocyte complexes and decreased fertilization rates
.
Endocrinology
2010
;
151
:
5438
5445
.

Wu
LL
,
Russell
DL
,
Norman
RJ
,
Robker
RL
.
Endoplasmic reticulum (ER) stress in cumulus-oocyte complexes impairs pentraxin-3 secretion, mitochondrial membrane potential (DeltaPsi m), and embryo development
.
Mol Endocrinol
2012
;
26
:
562
573
.

Wu
LL
,
Russell
DL
,
Wong
SL
,
Chen
M
,
Tsai
TS
,
St John
JC
,
Norman
RJ
,
Febbraio
MA
,
Carroll
J
,
Robker
RL
.
Mitochondrial dysfunction in oocytes of obese mothers: transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors
.
Development
2015
;
142
:
681
691
.

Yoshida
S
,
Kaido
M
,
Kitajima
TS
.
Inherent Instability of Correct Kinetochore-Microtubule Attachments during Meiosis I in Oocytes
.
Dev Cell
2015
;
33
:
589
602
.

Yuan
L
,
Liu
JG
,
Zhao
J
,
Brundell
E
,
Daneholt
B
,
Hoog
C
.
The murine SCP3 gene is required for synaptonemal complex assembly, chromosome synapsis, and male fertility
.
Mol Cell
2000
;
5
:
73
83
.

Yuan
L
,
Pelttari
J
,
Brundell
E
,
Bjorkroth
B
,
Zhao
J
,
Liu
JG
,
Brismar
H
,
Daneholt
B
,
Hoog
C
.
The synaptonemal complex protein SCP3 can form multistranded, cross-striated fibers in vivo
.
J Cell Biol
1998
;
142
:
331
339
.

Yun
Y
,
Holt
JE
,
Lane
SI
,
McLaughlin
EA
,
Merriman
JA
,
Jones
KT
.
Reduced ability to recover from spindle disruption and loss of kinetochore spindle assembly checkpoint proteins in oocytes from aged mice
.
Cell Cycle
2014
;
13
:
1938
1947
.

Zaragoza
MV
,
Jacobs
PA
,
James
RS
,
Rogan
P
,
Sherman
S
,
Hassold
T
.
Nondisjunction of human acrocentric chromosomes: studies of 432 trisomic fetuses and liveborns
.
Hum Genet
1994
;
94
:
411
417
.

Zhang
L
,
Cui
X
,
Schmitt
K
,
Hubert
R
,
Navidi
W
,
Arnheim
N
.
Whole genome amplification from a single cell: implications for genetic analysis
.
PNAS
1992
;
89
:
5847
5851
.

Zielinska
AP
,
Holubcova
Z
,
Blayney
M
,
Elder
K
,
Schuh
M
.
Sister kinetochore splitting and precocious disintegration of bivalents could explain the maternal age effect
.
eLife
2015
;
4
:
e11389
.

Author notes

These authors are contributed equally to the study.