ArticlePDF Available

Non-invasive oocyte quality assessment

Authors:
  • CHUV-Lausanne University Hospital

Abstract

Oocyte quality is perhaps the most important limiting factor in female fertility; however, the current methods of determining oocyte competence are only marginally capable of predicting a successful pregnancy. We aim to review the predictive value of non-invasive techniques for the assessment of human oocytes and their related cells and biofluids that pertain to their developmental competence. Investigation of the proteome, transcriptome, and hormonal makeup of follicular fluid, as well as cumulus-oocyte complexes are currently underway; however, prospective randomized non-selection-controlled trials of the future are needed before determining their prognostic value. The biological significance of polar body morphology and genetics are still unknown and the subject of debate. The predictive utility of zygotic viscoelasticity for embryo development has been demonstrated, but similar studies performed on oocytes have yet to be conducted. Metabolic profiling of culture media using human oocytes are also limited and may require integration of automated, high-throughput targeted metabolomic assessments in real time with microfluidic platforms. Light exposure to oocytes can be detrimental to subsequent development and utilization of time-lapse imaging and morphometrics of oocytes is wanting. Polarized light, Raman microspectroscopy, and coherent anti-Stokes Raman scattering are a few novel imaging tools that may play a more important role in future oocyte assessment. Ultimately, the integration of chemistry, genomics, microfluidics, microscopy, physics, and other biomedical engineering technologies into the basic studies of oocyte biology, and in testing and perfecting practical solutions of oocyte evaluation, are the future for non-invasive assessment of oocytes.
Received: October 5, 2021. Revised: January 4, 2022. Accepted: January 11, 2022
© The Author(s) 2022. Published by Oxford University Press on behalf of Society for the Study of Reproduction. All rights reserved. For permissions, please
e-mail: journals.permissions@oup.com
Biology of Reproduction, 2022, 117
https://doi.org/10.1093/biolre/ioac009
Oocyte Special Issue
Non-invasive oocyte quality assessment
Romualdo Sciorio1,Daniel Miranian2and Gary D. Smith2,3,*
1Edinburgh Assisted Conception Programme, EFREC, Royal Infirmary of Edinburgh, Edinburgh, UK
2Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
3Department of Physiology, Urology, and Reproductive Sciences Program, University of Michigan, Ann Arbor, MI, USA
*Correspondence: 4421 Medical Sciences I, 1301 E Catherine Street, Ann Arbor, MI 48109-5617, USA. Tel: +0017347644134; E-mail: smithgd@umich.edu
Abstract
Oocyte quality is perhaps the most important limiting factor in female fertility; however, the current methods of determining oocyte competence
are only marginally capable of predicting a successful pregnancy. We aim to review the predictive value of non-invasive techniques for the
assessment of human oocytes and their related cells and biofluids that pertain to their developmental competence. Investigation of the proteome,
transcriptome, and hormonal makeup of follicular fluid, as well as cumulus-oocyte complexes are currently underway; however, prospective
randomized non-selection-controlled trials of the future are needed before determining their prognostic value. The biological significance of
polar body morphology and genetics are still unknown and the subject of debate. The predictive utility of zygotic viscoelasticity for embryo
development has been demonstrated, but similar studies performed on oocytes have yet to be conducted. Metabolic profiling of culture media
using human oocytes are also limited and may require integration of automated, high-throughput targeted metabolomic assessments in real
time with microfluidic platforms. Light exposure to oocytes can be detrimental to subsequent development and utilization of time-lapse imaging
and morphometrics of oocytes is wanting. Polarized light, Raman microspectroscopy, and coherent anti-Stokes Raman scattering are a few novel
imaging tools that may play a more important role in future oocyte assessment. Ultimately, the integration of chemistry, genomics, microfluidics,
microscopy, physics, and other biomedical engineering technologies into the basic studies of oocyte biology, and in testing and perfecting practical
solutions of oocyte evaluation, are the future for non-invasive assessment of oocytes.
Summary Sentence
Direct and/or indirect non-invasive assessments of oocyte quality are reviewed and discussed in relation to clinical needs in assisted reproductive
technologies and as informative underpinnings of basic reproductive biology spanning follicle to embryo development.
Keywords: oocyte, oocyte maturation, chromatin content, non-invasive assessment, follicle, cumulus cells
Introduction
The oocyte morphological assessment represents a crucial step
in the assisted reproductive technologies (ART) laboratory
workday routine. Currently, it is generally very simple com-
pared to embryo and sperm assessment. Nonetheless, oocyte
quality is probably the most important limiting factor in
female fertility, playing a crucial role during the fertilization
process and subsequent embryo development [1]. According
to some authors [2], the phenotype of the adult stage offspring
is considerably correlated to the quality of the oocytes of ori-
gin. Indeed, the embryo is the result of the correct meeting of a
spermatozoon and an oocyte. The right selection of the oocyte
to be used for an in vitro fertilization (IVF) procedure would
be a logical tool for the reduction of the number of embryos to
be produced without changing the pregnancy outcomes. IVF is
a complex multistep procedure, the first of which is controlled
ovarian stimulation (COS). This process involves the use of
exogenous gonadotropins to stimulate the patient’s ovaries
to produce oocytes, which are then retrieved transvaginally
[3]. This application in ART further complicates the scenario.
Compared to the in vivo process, where oocyte maturation
occurs as the result of a long and natural follicle growth and
selection [4], common COS procedures suppress this natural
selection and allow successful maturation of oocytes that
otherwise would not develop spatially or temporally within
a pool of follicles. This may result in questionable and inher-
ently compromised quality, and could eventually be respon-
sible for fertilization failure, compromised embryo develop-
ment, or long-term consequences in offspring health [5].
It has been reported that even following intracytoplasmic
sperm injection (ICSI), human oocytes sometimes fail to fer-
tilize; those unfortunate events occur at an estimated rate of
3% [6]. Thus, this is a clear demonstration that successful
fertilization is a more complex process, related not only to
sperm penetration, but to many other processes inherent to
oocyte quality that are crucial and responsible for regulating
the majority of molecular and cellular mechanisms required
for successful fertilization. The concern is that a portion of
oocytes retrieved following COS is probably of compromised
quality; therefore, despite the optimal embryology condition
and efforts, they are eventually destined to fertilization failure,
or subpar embryo development, and/or the inability to estab-
lish a normal pregnancy. Furthermore, it is highly unlikely
that a single effector can account for these subpar outcomes.
The answer to these above queries, and many others in
reproductive biology, needs to be elucidated through a greater
understanding of regulatory mechanisms involved in oocyte
growth, maturation, and function [7].
We and others would propose that normal oocyte matu-
ration cannot be established simply by assessment of polar
body extrusion, but rather involves numerous cytoplasmic
processes not observed at the light microscope level. Those
mechanisms and signaling pathways in the oocyte cytoplasm
are vital for production and storage of carbohydrates and
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
2Non-invasive oocyte quality assessment
proteins, successful position of organelles, and regulation
of metabolic pathways required for oocyte maturation and
competence to support normal fertilization and embryo devel-
opment. In other words, the oocyte quality likely relies not
only on the nuclear and mitochondrial genome, but on the
microenvironment provided by the ovary, mainly during fol-
liculogenesis, and the preovulatory follicle. This multitude of
effectors can modify oocyte transcription and oocyte transla-
tion, and therefore negatively affect the development potential
of the future embryo [8,9]. In this scenario, comparable to a
big puzzle, the artificial environment of the IVF laboratory
plays only a small role. The purpose of this review is to
evaluate and synthesize the predictive value of morphological
features of mature human oocytes—and their related cells
and biofluids—to their developmental competence. Since the
simple morphologic evaluation, before and after fertilization,
may not be the best measurement for oocyte competence, we
will discuss features of the oocyte, its surrounding environ-
ment, and potential analytical markers that may be required
to support more complete information about oocyte quality
and subsequent embryonic developmental competence.
The importance of oocyte quality in ARTs
Every oocyte likely has a different developmental potential,
and successfully predicting that potential is of great interest
for ART practitioners for a multitude of reasons. The
ability of an oocyte to undergo meiotic maturation has
been termed meiotic competence, and the ability to go on
to support successful embryonic development is defined as
developmental competence. In combination, these constitute
oocyte competence [10]. While maternal age is the best
predictor of oocyte competence [11], variation exists between
the competency of different oocytes retrieved during the same
COS, and currently, the most reliable methods of determining
oocyte competence are only marginally capable of predicting
a successful pregnancy [12]. Some of the most obvious
benefits of better predictive assessment of oocyte competence
are higher live pregnancy rates and fewer early pregnancy
losses [13]. An improved ability to predict oocyte potential
is also essential to the widespread adoption of elective single
embryo transfer (eSET), further reducing the morbidity and
higher costs of multiple gestations associated with multiple
embryos transferred [14]. Additionally, it is conceivable
that as the ability to predict oocyte competence continues
to improve, stimulation protocols could be optimized to
select for quality over quantity of oocytes retrieved, making
the experience more patient-friendly, with fewer cases of
ovarian hyperstimulation syndrome, and without sacrificing
outcomes [15]. The ability to forecast pregnancy outcomes
before oocytes are fertilized is also desirable, as some patients
are opposed to the creation of unused embryos and certain
countries have restrictive laws regarding embryo production
[13,16]. The growing cost associated with freezing additional
embryos could also be mitigated. Finally, improved non-
invasive assessment would allow better counseling to women
undergoing oocyte cryopreservation in the setting of fertility
preservation or for elective reasons [17].
Current oocyte nuclear maturation
assessment in relation to ARTs
In contemporary ART involving ICSI, the cumulus oocyte
complex (COC) must be denuded in order to remove all the
cells surrounding the oocyte, thus allowing visual assessment
of the sperm injecting procedure. The first step consists of
exposure to the hyaluronidase enzyme, followed by mechan-
ical force applied using pipettes of a diameter slightly larger
than the oocyte and surrounding cumulus. This allows accu-
rate assessment of oocyte nuclear maturation [GV-intact (GV-
I), metaphase I/anaphase I/telophase I (collectively termed MI)
or metaphase of meiosis II (MII)]. The assessment by light
microscopy can display the presence of the first polar body
(PBI) in the perivitelline space (PVS), which is considered to be
a marker of nuclear maturation. Oocytes with clear extrusion
of the PBI are supposedly at MII, with the chromatin aligned
on the equatorial plate of the meiosis II metaphase spindle.
However, recent studies using polarized light microscopy have
shown that a small percentage of oocytes displaying the PBI
in the PVS may still be immature at the telophase of meiosis I
stage [18]. In this case, a true oocyte maturation at MII might
be confirmed by the presence of the meiotic spindle (MS),
which can be localized underneath the PBI [19,20] and will
be discussed further below in relation to the use of polarized
microscopy. Additional studies have also examined the MS
and the orientation to the PBI to be used as a prognostic
tool. Oocytes with the MS separated >90from the PBI have
lower fertilization rates compared to oocytes where the MS
is oriented <90from the PBI [18]. However, a considerable
number of studies have analyzed the importance of the MS
orientation in human oocytes, and its presence has been
associated with fertilization rates and pregnancy outcomes
with generally contradictory results [2127]. A contemporary
review on MS assembly, positioning, and function is avail-
able [28]. Nonetheless, it is important to consider that the
daily routine work in the embryology laboratory, including
manipulation and culture conditions, might have an effect
on the cytoskeleton of the oocyte, compromising the MS
and eventually being responsible for lower fertilization rates
[29]. However, following COS, it is generally expected that
around 80–85% of the oocytes are at the MII stage, with
clear extrusion of the PBI, whereas another 5–10% are at
GV-I stage and can be recognized by the spherical nucleus
containing a large exocentric nucleolus typical of the prophase
of meiosis I. Another 5–10% of oocytes with the absence of
both PBI and GV are classified as being at MI. These oocytes
have undergone GV breakdown, but have not fully pro-
gressed through meiosis I and are developmentally somewhere
between MI and MII, where the chromosomes are aligned on
the metaphase plate in preparation for finishing the first meio-
sis division [30]. The critical importance of proper chromatin
segregation/regulation during nuclear maturation and oocyte
cytoplasmic maturation in yielding oocytes with developmen-
tal competence has been reviewed elsewhere [3133].
Defining non-invasive oocyte assessment and
its needs in ARTs
In IVF treatment, the selection of a single embryo to transfer
often presents a challenge to the embryology team, consider-
ing limited information available on an embryo’s viability. A
relevant percentage of embryos still fail to implant, despite
being morphologically scored as “top quality embryos,” or
even after having been genetically analyzed. As mentioned
earlier, the goal of an IVF cycle should be only one healthy
baby born per transfer; therefore, eSET should be regularly
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 3
employed in order to avoid the risk of prematurity and compli-
cations associated with multiple births [34]. The need to estab-
lish a non-invasive method to identify specific characteristics
of oocytes and embryos that reflect normal health function
or the ability to further develop into a healthy pregnancy is
wanting. To be non-invasive, a technology should have zero
detrimental impact on the oocyte, its fertilization, subsequent
embryo development, ability to establish a pregnancy, and a
healthy offspring with an expectation of a healthy lifespan
(similar to non-ART-conceived individuals). Going forward,
we will continue to discuss the current light microscope
non-invasive assessment of the oocyte. We will also consider
assessment of oocyte developmental competences that may be
invasive today, yet provide informative pathways forward that
may be non-invasive in the future. Finally, we will also discuss
non-invasive assessments that are making progress toward
utility in oocyte assessment.
The light microscopic appearance of the cytoplasm has been
the subject of several published studies that try to establish
its correlation with fertilization and pregnancy outcomes.
The definition of features differed between studies; there-
fore, relative analysis of results is difficult. Some authors
analyzed and defined the feature as “dark cytoplasm” [35,
36], whereas others used “diffused cytoplasmic granularity”
[37], “dark granular appearance of the cytoplasm” [38], or
“dark cytoplasm with slight granulation” [39]. When this
feature was analyzed and attempts were made to establish
a correlation with pregnancy outcomes, some authors found
that “dark cytoplasm” alone was not a predictive factor in
“in vitro” or “in vivo” parameters [35,38,39], whereas
others reported that embryo quality was compromised when
embryos developed from oocytes with dark cytoplasm [36,
40]. However, Wilding and colleagues [41] reported that cyto-
plasmic granulation was associated with higher fertilization
rates compared to oocytes with a total absence of granularity.
Therefore, the debate on dark cytoplasm and cytoplasmic
granulation is still alive, yet one can appreciate that these
observations are subjective and likely lack concordance within
and between laboratories.
Another recently investigated aspect is the relationship
between oocyte/embryo function and their biomechanical or
biophysical properties [42]. The viscosity of the cytoplasm
and the resistance of the cell membrane at ICSI were
analyzed by Ebner and collaborators [43] and Wilding
and coauthors [41]. Both viscosity and resistance had a
significant effect on outcomes such as fertilization, embryo
quality, and blastocyst formation. These cytoplasmic viscosity
differences have also been correlated with patients’ infertility
features and with ovarian stimulation regimens [43,44]. It
has been also reported that during follicle maturation and
COS, the oocyte cytoplasm viscosity might undergo several
changes—from an aqueous to a more viscous and sticky
substance. In the COS cycle that results in hyperstimulation,
the oocyte cytoplasmic maturation process often seems to
be out of sync when compared to nuclear maturation [43,
44]. In addition, viscosity in mature oocytes has also been
investigated, with findings revealing that higher viscosity
might be a poor prognostic indication of embryo development
and implantation [43]. Viscoelasticity in the cytoplasm of
mature oocytes and membrane resistance may be another
important feature to take into consideration, and perhaps
clarifying why some oocyte membranes break rapidly
at ICSI sperm injection, whereas those of other oocytes
fail to break even with aspiration [45]. Specific reports
have recently been published assessing viscoelasticity of
the oocyte and zygote as potential predictors of embryo
development [46] and will be discussed in further detail
below.
Non-invasive assessment of the follicular
environment and polar bodies
Analyzing follicular fluid (FF) is an appealing option for non-
invasive assessment of oocyte competence, as it is collected
during every oocyte retrial and is usually discarded. FF is
composed of granulosa and theca cell secretory products, as
well as the blood plasma components that cross the blood
follicle barrier [47]. Additionally, the FF is in intimate contact
with the maturing oocyte, and it is hypothesized that it may
contain evidence of either healthy or unhealthy developmental
conditions. Because the FF is likely different between oocytes
even during the same COS, one must be careful to only study
the associations between oocytes and their respective FF. This
creates some technical challenges, as increased needle sticks
during transvaginal oocyte pick up put the patient at increased
risk of bleeding, and the inherent dead space in the aspiration
tubing requires a certain amount of the buffer solution to be
wasted during each analysis. As a result, most FF studies have
looked at only the first oocyte retrieved from each ovary.
Follicular fluid transcriptomic as an indirect
non-invasive assessment of oocyte quality
Follicular fluid assessment has been categorized into the fol-
lowing groups for the purpose of this review: RNAs; pro-
teins (growth factors, anti-apoptotic factors, peptides, and
amino acids); hormones; reactive oxygen species; sugars; and
prostanoids. The term transcriptome indicates the total RNA
content inside a cell/biosample, including messenger RNA
(mRNAs), ribosomal RNAs, transfer RNAs, and microRNAs.
Analyzing the FF transcriptome is particularly challenging for
several reasons [48]. There are over 5000 genes known to be
expressed on the human oocyte [49] and the sheer number
of specific mRNA expressed by the oocytes may be likely
twice that number [50]. Additionally, because the FF also
contains mRNA from surrounding granulosa cells, providing
predictive information regarding oocyte potential from the
FF transcriptome using current techniques is limited. Since
mRNA is ultimately translated into proteins that have the
biggest impact on cell functionality and metabolism, analysis
of FF proteins (proteomics) or metabolites (metabolomics) has
proven to be more fruitful [48]. With this said, studies of
non-translated RNAs, such as long non-coding RNAs [51]are
beginning to be reported and may also have future predictive
value of oocyte developmental competence.
Follicular fluid proteomic as an indirect
non-invasive assessment of oocyte quality
One study analyzed the FF of healthy ovum donors using high-
pressure liquid chromatography (LC) at differing pH levels,
followed by mass spectrometry (MS) to separate a total of
742 proteins. This same study also used qualitative analysis to
compare samples of FF from aged matched controls of infertile
women undergoing IVF pre- and post-hCG administration,
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
4Non-invasive oocyte quality assessment
and found 11 proteins that were significantly higher in the
pre-hCG samples and 6 proteins that were significantly lower
in the post-hCG samples. These proteins belonged to sev-
eral different functional groups, including protease inhibitors,
inflammation-mediated proteins, and cell adhesion proteins
[52]. Another study looked at the FF of three women with
recurrent pregnancy loss (RPL) and compared it to the FF
of three control women who had a history of at least two
prior births and no miscarriages. They found that the FF
of women with RPL had significantly less expression of
five different proteins, notably complement component C3c
chain E, fibrinogen γ, antithrombin, angiotensinogen, and
hemopexin precursor [53]. A different study looked at the
FF of women grouped as having either advanced maternal
age (AMA), a history of implantation failure, or AMA plus
a history of implantation failure, to see if there was an
association with polar body aneuploidy. They analyzed the
levels of FSH, LH, progesterone (P4), estradiol (E2), and Anti-
Müllerian hormone (AMH) and found that high FF FSH
levels were associated with PB aneuploidy. This may not be
surprising, however, there has been mixed evidence to date as
to whether the high doses of gonadotropins required for COS
lead to aneuploidy or not [5461]. The other FF components
tested had no predictive value in aneuploidy, which is notable
because AMH FF has previously been shown to be a marker of
successful oocyte fertilization [62]. Because of the lack of clear
prognostic value when only one FF marker is used, some inves-
tigators have recently turned to analyzing a panel of markers
simultaneously. Recently, FF AMH and P4 levels of oocytes
that had been successfully fertilized using ICSI were both
analyzed together and grouped based on whether or not they
successfully progressed to the blastocyst stage. When looked
at in isolation, having a FF AMH level >15 pmol/L or a FF P4
level >60 mg/mL corresponded to a positive predictive value
of blastocyst formation of 76.96% and 90.99%, respectively.
However, when these same FF cutoffs were used in con-
junction, the positive predictive value of successful blastocyst
formation rose to 96.83% [63]. This was a relatively small
cohort of 88 patients and will need to be replicated, but it
again demonstrates the potential usefulness of FF proteomics.
Follicular fluid steroids and fatty acids as an
indirect non-invasive assessment of oocyte
quality
Steroids such as estrogen and testosterone in conjunction with
gonadotropins are believed to influence oocyte maturation,
but their exact role needs to be investigated further. It has been
shown that priming with estrogen is necessary for Ca2+oscil-
lations to occur during maturation, but improper estrogen-
to-testosterone ratios can lead to an abnormal Ca2+response,
thus negatively impacting embryo development [64]. Estrogen
(E2) and P4 within FF have been the target of investigation
for implications on pregnancy outcomes. It has been well-
documented that elevated FF E2 and an increased FF E2/P4
ratio are associated with a higher chance of achieving a
pregnancy [6573]; however, there have been other stud-
ies that have not confirmed these findings [7478]. Dehy-
droepiandrosterone (DHEA), is converted to testosterone by
theca cells in ovarian connective tissue [79]. The sulfatase
enzyme SULT2A1 reversibly catalyzes DHEA to its sulfate,
DHEA-S, which has been shown to be the precursor to
estrogen production in granulosa cells [80]. In fact, DHEA-
S is found in micromolar concentration in the FF, making
it the most abundant steroid in FF [81]. The interconver-
sion between DHEA and DHEA-S and their function in
both ovarian androgen and testosterone production has made
them logical targets for evaluation during IVF cycles. Interest-
ingly, initial studies looking at serum DHEA-S levels during
COS found no association with successful pregnancies [82],
whereas high DHEA levels in the FF were shown to have a neg-
ative correlation to oocyte retrieval number and subsequent
fertilization [83]. However, a more recent study looked at FF
DHEA-S and E2 levels of 521 women undergoing IVF. When
these women were stratified by low, medium, and high FF
DHEA-S, statistically higher pregnancy rates were seen in the
high FF DHEA-S cohort. There were also significantly more
total fertilization failures seen in the low FF DHEA-S cohort
compared to the medium and high FF DHEA-S groups. Raised
FF E2 levels were also seen in the high FF DHEA-S group,
as well as among the low and medium FF DHEA-S patients
who achieved pregnancy [84]. These findings corroborate the
role of estrogen in oocyte priming and suggest DHEA-S, as
a precursor to estrogen, may play an important role in this
mechanism.
Investigators have also looked to other steroid-related
molecules found in FF as potential prognostic markers. A
study conducted using the FF of the lead follicle of 54
women undergoing IVF found that oocytes that did not
show two pronuclei or underwent degeneration after ICSI
had significantly higher levels of arachidonic acid and linoleic
acid derivatives, as well as increased activity of secretory
phospholipase A. This study used strict selection criteria to
omit women over the age of 37, with BMIs 26, affected by
male factor infertility, or with poor IVF response [15]. These
fatty acids have been implicated in inflammatory activators
that may have an impact on the developing oocyte [85];
therefore, the selection criteria in the study was careful to
exclude conditions that may cause a baseline alteration in
inflammatory response or steroid levels. It is worth noting that
many of the studies excluded certain subsets of the infertile
population in their initial analysis, and larger studies including
more diverse patient populations will be needed to decipher
variations among different diseases and patient demographics
before these techniques can be reliably employed on a wide
scale.
Cumulus cells transcriptomics, metabolites,
and proteomics as indirect non-invasive
assessment of oocyte quality
Transcriptomic approaches can be used to explore the gene
expression level of cumulus and granulosa cells, in somatic
cells of the follicle, and in the embryos after fertilization [86].
The transcriptomics technology might help to determine new
diagnostic features indicative of oocyte and/or early embryo
development. Currently, assays of single gene activity can be
performed and associated with oocyte maturation, fertiliza-
tion, embryo competence, and future pregnancy outcomes.
Recent studies suggest that the individual transcript levels
of specific genes expressed in the COC are well-correlated
with oocyte maturation [87,88], embryo viability [8790],
ongoing pregnancy [9092], or live birth outcome [90,93].
However, it needs to be mentioned that the collection of cells
from the COC, although considered non-invasive, requires
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 5
the oocyte to be exposed to additional time outside the incu-
bator, which can be detrimental to subsequent development.
In addition, single COC culture needs to be performed in
order to retain the identity of each oocyte sample and its
resulting embryo, which can add extra cost and workload.
Nonetheless, the studies reported earlier are performed with
retrospective designs, without any clinical validation of the
identified markers. Further studies need to be completed with
the goal of identifying novel markers in the IVF setting that
will correlate to pregnancy outcomes [94].
The functional well-being of granulosa cells and/or cumulus
cells might be reflected in the oocyte quality [95,96]. During
the meiotic arrest, cyclic guanosine monophosphate from
cumulus cells goes into the oocyte through gap junctions and
impedes the hydrolysis of cyclic adenosine monophosphate
(cAMP) by the phosphodiesterase PDE3A. This inhibition
maintains a high concentration of cAMP in the oocyte and
keeps meiosis arrested. The preovulatory LH induces the
reduced levels of intra-oocyte cAMP and meiosis resumption
[9799]. Growth differentiation factor 9 is a protein that, in
humans, is synthesized by the oocyte and plays an important
role in the expansion of cumulus cells, which is a critical aspect
of final follicle development [100,101]. Oocytes collected
from follicles with reduced expansion have a lower chance
of producing a good quality embryo and limited potential
for implantation (review: [102]). McReynolds and cowork-
ers [103] evaluated both the transcriptome and proteome
of human cumulus cells from young fertile oocyte donors
and infertile women of AMA and found striking differences
at the protein level. These investigators were able to iden-
tify 1423 cumulus cell proteins. When comparisons were
made between the abovementioned groups, statistical analysis
revealed 110 (7.7%) proteins to be differentially expressed.
Pathway annotation revealed significant involvement of pro-
teins in metabolism, oxidative phosphorylation, and post-
transcriptional mechanisms relative to AMA. In addition, gene
expression analysis showed altered profiles in cumulus cells
from women in their early to mid-40s. These types of analyses
are indicative of non-invasive assessment of oocyte-associated
transcripts and proteins that may have practical application in
the future. Prospective randomized non-selection-controlled
trials of the future will inform as to the reliability of these
cumulus transcripts and proteins as having a prognostic value
in determining oocyte developmental competence.
Polar body morphology and genetics as
indirect non-invasive assessments
of oocyte quality
The PBI is normally present in the PVS and is generally
smooth and without fragmentation. However, the impact and
the biological significance of PBI morphology, dysmorphisms,
or fragmentation are still unknown and a subject of debate.
Fragmented PBI should not be particularly considered an
oocyte marker, since the fragmentation may be related to the
post-ovulatory time. However, it has been suggested that a
degenerated PBI might be associated with asynchrony between
nuclear and cytoplasmic maturation, potentially related to
overmaturity of the oocytes [104]. Oocytes displaying a clear
and intact PBI have increased ability of producing blastocysts
and higher pregnancy rates [105,106]. Several studies have
been performed with the goal of determining the relationship
between PBI morphology and ICSI outcome, but were unable
to find a clear link between the two features [107,108].
Exceedingly large PBI seems to be a sign of poor prognosis
and is associated with compromised embryo viability [107,
109]. In addition, large PBI is related to high frequencies of
multinucleated blastomeres and might therefore contribute to
embryo aneuploidies [107].
It has been reported that the majority of aneuploidies in
human preimplantation embryos are derived from meiotic
errors occurring during oogenesis due to the lack of effi-
cient molecular mechanisms to repair eventual errors dur-
ing the meiotic division [110,111]. In addition, a growing
body of evidence suggests that environmental contaminants,
endocrine-disrupting chemicals, and air pollution, are posing
major threats to human reproductive health [112]. In that con-
text, preimplantation genetic testing for aneuploidies (PGT-A)
was introduced a few decades ago with the aim of identifying
euploid embryos to be transferred in IVF cycles [113]. Polar
body biopsy is an alternative technique to cleavage stage or
blastocyst biopsy first introduced by Verlinsky and colleagues
[114]. The main disadvantage of the PB biopsy technique is
that only the maternal aneuploidies can be identified; it will
not detect paternal meiotic or post-zygotic mitotic errors. A
considerable benefit of PB biopsy is the greater time available
to perform genetic testing without requiring embryo cryop-
reservation. An additional benefit is that PB biopsy avoids
embryo manipulation, which might be important in countries
where, due to ethical concerns, the manipulation of human
embryos is prohibited. In 2012, the European Society of
Human Reproduction and Embryology initiated a multina-
tional, multicenter, randomized clinical trial to assess the effec-
tiveness of PGT-A performed with PB biopsy by microarray
analysis. The main question of the study, named ESTEEM, was
to evaluate whether the analysis of 23 chromosomes in the PBI
and PBII and the selection of a euploid embryo for transfer
increased the chance of a live birth within one year among
women aged 36–40 years compared to ICSI without chro-
mosome analysis. Between June 2012 and December 2016,
205 consenting women were assigned to ICSI treatment with
PGT-A (study group) and 191 to ICSI without PGT-A (control
group) [115]. The results reported that the proportion of
women with a live birth within one cycle, followed by delivery,
was not significantly different between the two groups: 50
of the 205 with chromosome screening (24%) versus 45 of
191 without chromosome screening (24%). However, genetic
testing platforms are continually evolving, with microarray
analysis now being replaced by next-generation sequencing
(NGS), single-cell NGS, and non-invasive NGS.
Different methods have been proposed to identify aneuploi-
dies of all chromosomes in ARTs. Whole genome amplifica-
tion amplifies the genome and therefore results in an adequate
starting material for comprehensive chromosome screening
[116]. Other methods are metaphase comparative genomic
hybridization [117], array comparative genomic hybridization
[118], genome-wide single nucleotide polymorphism analysis
[119], PCR-based detection [120], and NGS [121], which is
becoming the procedure of choice for PGT-A, with improved
results compared to a-CGH [122]. In theory, although the
introduction of new molecular technologies should be able
to provide a better resolution for distinguishing aneuploidies,
there are some authors who have reported that those methods
may be intrinsically imperfect and require further optimiza-
tion [123]. Additionally, due to the small amount of DNA
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
6Non-invasive oocyte quality assessment
collected from PBI, errors might occur more frequently during
the genetic analysis. Published studies reported a poor consis-
tency between the molecular analyses performed in different
laboratories, reflecting the potential of different molecular
technologies giving different interpretive information [124,
125]. It also must be noted that in most of these clinical PGT-
A analyses, the results are based on a single observation, an
n=1. This is concerning when one considers the standard of
care relating to clinical assays, be it chemistry, molecular biol-
ogy, microbiology, or genetics. Whether the genetic assessment
of the oocyte polar body with these newer platforms will yield
useful, practical, and informative selective power of oocyte
developmental competence is yet to be realized.
Non-invasive assessment of oocytes as a
direct evaluation of oocyte quality
Oocyte biomechanical properties
The study of biomechanical properties of cells and their result-
ing health, function, and development is becoming increas-
ingly appreciated. In both stem cells and ovarian cancer cells,
viscoelasticity has been shown to influence cell structure,
function, and biophysical properties [42,126]. Viscoelasticity
is defined as the property of materials that exhibit both vis-
cous and elastic characteristics when undergoing deformation.
Cell membranes are an example of a viscoelastic material
[127,128], and intracellular cytoskeletal organization and
remodeling have important relations to a cell’s viscoelastic
nature [129]. Studies have demonstrated that interactive bio-
physical forces, such as cell/membrane deformation and fluid-
flow shear stresses, impact cellular viscoelasticity [130132].
It has been proposed that cell function and biomechanical
changes are a result of viscoelastic intracellular signaling
through kinase/phosphatase–structural cytoskeleton interac-
tions [133136] and/or intracellular Ca2+[137]. While the
majority of these viscoelastic studies have been performed
in non-gamete cells, there are recent and significant reports
suggesting that measurements of oocyte biomechanical prop-
erties may provide insight into oocyte and subsequent embryo
function and development.
One oocyte-associated structure that may have significant
assessment discernment is the zona pellucida (ZP). The mam-
malian ZP is a sulfated glycoprotein matrix that is assembled
surrounding the primary oocyte during the early stages of
folliculogenesis [138]. The mouse ZP is composed of three
ZP glycoproteins, termed ZP1, ZP2, and ZP3 [139,140],
whereas the human ZP has four glycoproteins [141,142]. The
ZP serves as a molecular “gatekeeper” by providing species
specificity during binding of the spermatozoa to the oocyte
[143], stimulates the spermatozoa acrosome reaction [144],
and, following single-sperm binding and passage, is modified
to function as a component of the block to polyspermia [145,
146]. Using atomic force spectroscopy, Papi and colleagues
demonstrated that the ZP of immature eggs has a pure elas-
tic behavior, whereas in mature and fertilized oocytes, the
ZP transitions to a more plastic behavior—with a higher
force required to induce deformation [147]. It has been well-
recognized that following fertilization, the release of oocyte
cortical granule enzymatic contents into the PVS results in ZP
“hardening”—defined by both relative resistances to exoge-
nous proteolytic exposure (decreased solubility) [148] and/or
nanometrically measured mechanical properties [149]. Drob-
nis and coworkers performed early studies on direct measure-
ments of ZP “hardening” as a mechanical end-point measure.
These studies provided a much-needed understanding of the
relations of solubility and mechanical changes, as well as
a delineation of these two measures in relation to oocyte
maturity, fertilization, and species differences.
Independent of oocyte development, maturation, and/or
fertilization differences in ZP structure/function, the human
oocyte ZP’s association with embryonic developmental com-
petence and subsequent pregnancy success has also been inves-
tigated. Early works focused on ZP thickness measures and
ZP thickness variations across its two observed dimensions
[150,151]. Both of these studies reported that ZP thickness
variations of >20–25% were positively predictive of preg-
nancy success. In addition, birefringent microscopy, which
enables visualization of the inner, middle, and outer zones of
the ZP, has supported the idea that magnitude of light retar-
dance of the ZP inner layers may be a non-invasive measure
of oocyte developmental competence [44,152]. Collectively,
these retrospective, non-selective studies of ZP measures and
subsequent embryo development and/or pregnancy success
are encouraging as potential non-invasive oocyte assessments;
however, properly powered prospective randomized trials are
wanting.
Elegant studies performed by Yanez and colleagues have
demonstrated the ability to measure viscoelastic properties
on mouse and human zygotes [46] using a micropipette aspi-
ration platform that allowed quantification of the depth of
aspiration of the ZP, and part of the cell membrane, over
time. These investigators reported the ability to quantify four
biomechanical parameters of a linear elastic solid model for
each zygote measured. While no individual parameter had pre-
dictive value in relation to subsequent embryo development,
combining parameters showed partitioning of zygotes des-
tined for blastocyst development [17,46]. While these studies
are promising and thought-provoking, such studies remain to
be performed on oocytes before fertilization. Additionally, as
mentioned above, prospective randomized controlled human
studies are needed to decipher if these viscoelastic measures
are predictive of developmental competence and pregnancy
success, and if they are accurately defined as non-invasive.
With that said, these and other performed and proposed
studies that integrate biomechanics, single-cell analysis, novel
means of microscopy (see: following sections), microfluidics,
and oocyte developmental biology likely will have research
and practical utility as non-invasive means of assessing and
quantifying oocyte developmental competence (review see:
[17,153,154]).
Oocyte metabolomics
Metabolomics is classically defined as the non-targeted
identification—and, importantly, quantification—of all low
molecular weight end products of metabolism (metabo-
lites) [155]. As the number of metabolites are reduced
approximately, two magnitudes reduced in comparison to
genomic and transcriptomic measures, it has been proposed
that metabolomic analysis can be performed more rapidly
[48]. The goals of metabolomics are to quantify either
individualized metabolites as biomarkers, or metabolic
fingerprints (grouping of multiple metabolites), in a cell
system under comparative physiological conditions at a given
developmental temporal set-point [156]. Two important
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 7
concepts make metabolomics attractive as a non-invasive
assessment of cellular function. First, it has been proposed
that cellular health can be delineated by the modified
concentrations of metabolites reflective of attempts to
maintain metabolic homeostasis [157]. Second, the products
of intracellular metabolism—the resulting metabolites—are
in dynamic balance with those in biofluids surrounding
the cells, whether those fluids represent plasma, FF, or
culture media [155]. For these reasons, and others men-
tioned below, the presumption that metabolic profiling of
individual or groupings of metabolites in culture media
of oocytes and/or embryos in ART might provide non-
invasive measures of cellular health, developmental compe-
tence, and selective advantages has been acknowledged for
decades.
Platforms for metabolomic analysis are many and can be
categorized as nuclear magnetic resonance techniques, MS
techniques, and vibrational spectroscopy. Mass spectrometry
can be used alone or with other chromatographic analyzes,
including liquid chromatography, ultra performance liquid
chromatography, gas chromatography (GC), GC-time of
flight (TOF), capillary electrophoresis, and Fourier transform-
ion cyclotron resonance. Extensive review of methods,
strengths and limitations, and applications are available
in the following reviews [158,159]. Independent of the
methodology used in metabolomics analysis, there are
critical components to the process that can directly influence
the fidelity of outcomes and variability of results. First,
metabolism must be halted in biological samples and/or
biofluids at comparable times and as rapidly as possible
to terminate metabolic and enzymatic activities. This can
be accomplished with snap-freezing. Metabolites can be
isolated by numerous procedures that each have advantages
and disadvantages [160]. Platforms used for metabolic
assessments need to be unbiased, sensitive, selective, and
compatible with high-throughput analysis, while resulting
large data sets require powerful bioinformatics and statistics
that are amendable to providing adequate data analysis and
appropriate interpretations [156].
There have been numerous targeted and non-targeted
metabolomics approaches to address domestic animal
nutrient and seasonal impact on gamete/embryo metabolism,
metabolism during development, and metabolic requirements
for development [161165]. While these studies have been
hypothesis-driven or focused on elucidating mechanistic
pathways in development, they have also given rise to tar-
geted—and to a greater extent, non-targeted—metabolomics
studies that attempt to predict oocyte/embryo quality. To date,
the majority of metabolomics studies related to predicting
and improving outcomes in human ART have been focused
on the preimplantation embryo. Although not the focus of
this paper, some of the outcomes of these human embryo
studies are informative about opportunities, as well as
limitations.
Numerous pilot, cohort, and case-controlled studies
have been performed regarding human preimplantation
embryo culture media metabolomics in predicting embryo
development, morphology, implantation, and successful
pregnancy [166173]. As one might anticipate these reports
provided positive correlations, no correlations, and ultimately
collective non-conclusive guidance for the utility of embryo
culture media metabolomics in predicting ART outcomes.
There have been three randomized controlled trials (RCTs)
[174176], which collectively concluded that metabolomics
(as performed in the trials) did not provide an objective
marker of embryo viability, did not demonstrate a benefit
in predicting a viable pregnancy, and did not significantly
improve selection of embryos in relation to live-birth rates
compared to embryo morphology selection alone. For more
detail on human embryo metabolic studies, one should
evaluate the review by Bracewell-Milnes and coworkers
[177]. The goals of this systematic review were to interrogate
published reports regarding metabolomics and human female
reproduction, with a specif ic focus on potential applications of
metabolomics of FF, embryo culture media, and endometrial
fluids in ART success.
To our knowledge, there is only a single peer-reviewed and
published report involving metabolomics profiling of human
oocytes [8]. The objectives of this study were to assess whether
near-infrared spectroscopy-generated metabolomic data col-
lected from short-term oocyte culture would correlate with
oocyte nuclear maturation and subsequent preimplantation
embryo development. In this case-controlled study, a total
of 412 oocytes from 43 patient ART cycles were denuded
of cumulus cell and placed into individual microdrops for
3–4 h prior to ICSI. Oocytes had different maturity (some
MI and some MII) and were followed in relation to day 3
and day 5 embryo development. The study authors concluded
that metabolomic profiling of spent media following short-
term culture of human oocytes can be performed, and is
(1) related to nuclear maturity, (2) able to predict embryo
development at both day 3 and day 5 stages, and (3) relates to
embryo viability. It is important to note that this study, like
many of the abovementioned human embryo studies, used
non-targeted metabolomics to gain metabolite fingerprints
with a computational categorization of profiles into “viability
indices” that may not necessarily be grounded in foundational
biology, development, and evidence-based predictions of cell
metabolism.
There are numerous extrinsic factors that can make
non-invasive assessment of the oocyte problematic. These
include non-oocyte cellular contamination, short culture
times, uncertainty of consistency of culture media and
media additives, the inf luence of interactive parameters
such as gaseous phase of culture, and human error inherent
to manual manipulations. While this study is interesting
and encouraging, additional studies are needed—especially
prospective RCTs. The above discussion also underscores
the need for well-controlled studies—not just in non-
targeted metabolomics, but also in targeted metabolomics.
Identification of predictive biomarkers of normal and
abnormal metabolism as it relates to proper and aberrant
human oocyte development and support of expected embryo
development is also wanting. Technical advances are being
explored in high-coverage targeted metabolomics [178]that
will likely advance the discovery of metabolic biomarkers.
The integration of automated, high-throughput targeted
metabolic assessments in real time, with microfluidics
platforms that are compatible with oocyte/embryo culture,
will be important in removing human technical variability
and standardizing assays and results. Multiple groups have
already demonstrated the ability to perform single embryo,
real-time metabolic assays assisted by microfluidic automated
fluid handling (Figure 1)[179,180]. It is quite easy to
recognize that multiple challenges exist in the application
of non-invasive oocyte metabolomics to predict ART
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
8Non-invasive oocyte quality assessment
Figure 1. A Braille display-based microfluidic embryo culture and assay system that allows live-cell assessment of glucose consumption and lactate
production in real time. (A) Photograph of the whole system placed on a microscope. (B) Schematic cross-section of the cartridge on microscope. (C)
Schematic design of the embryo culture and metabolism assay cartridge. ITO (indium-tin-oxide heater is attached to the Braille displayed module to
maintain the media temperature at approximately 37C. (D) Graphic representation of real time on-cartridge measures of controls (media with 100, 50,
and 0 μM glucose) demonstrating the ability to quantify fluid glucose in real time on-cartridge with measure obtained every 5 s and changing
concentrate every 24 min. (E) Not only was glucose consumption measured, but lactate production could be measured (picomoles/h). Lactate
production was measured from individual 2-cell mouse embryos (N=22) and individual mouse blastocyst (N=25) and levels measured in this
non-invasive manner, in real time on-cartridge, was similar to levels measured in the past with fluorescent microscopy (considered to be invasive if
performed on-cartridge). Figure adapted from [180] with permission from the Lab Chip, The Royal Academy of Chemistry.
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 9
outcomes, and even easier to accept the fact that advances in
many fields (biological and non-biological) and collaborative
integrations of techniques and expertise are needed to make
this vision come to fruition.
Oocyte novel imaging
Light microscopy and time-lapse imaging
The use of light microscopy is paramount to gamete and
preimplantation embryo assessment in the research and clini-
cal laboratory. The use of dissecting microscopes to find COCs
at retrieval is invaluable. Additionally, inverted phase-contrast
microscopy with Hoffman modulation or Nomarski differ-
ential interference allows for higher magnification and depth
of observation when assessing oocyte maturity, pronuclei for-
mation, and preimplantation embryo development. Recently,
time-lapse imaging of oocytes, zygotes, and embryos has been
investigated as a potential benefit in embryo selection by
morphometrics. Meseguer and coworkers demonstrated that
use of a time-lapse image acquisition system allowed determi-
nation of developmental events like the timing of cleavage and
suggested that using a multivariable model one could classify
human embryos with greater implantation potential [181].
Wong and colleagues reported that using non-invasive time-
lapse imaging of human embryos, one could predict blastocyst
development with developmental algorithms [182]. Chavez
and colleagues reported that similar time-lapse imaging sys-
tems could provide details in length of cell cycle progres-
sion and individual blastomere fragmentation that would be
predictive of embryo euploidy/aneuploidy [183]. While these
initial reports were exciting, subsequent larger, prospective,
and controlled studies have not necessarily confirmed such
findings. More recent reviews have concluded that evidence
does not support the routine use of time-lapse technology in
clinical ART laboratories and that future studies are needed
[184,185].
While the future use of time-lapse microscopy to assess
embryo developmental events and produce algorithms that
have predictive power remains a realistic research goal,
the use of time-lapse imaging or morphometrics as a non-
invasive microscopic imaging system for oocytes is wanting.
Beyond the phase contrast, optical imaging of oocytes without
cumulus cells to assess maturity (germinal vesicle intact,
metaphase I, or metaphase II), the utility of time-lapse
imaging of oocyte prior to insemination/fertility assessment is
questionable. Therefore, the evaluation of other microscopic
tools for oocyte assessment is required to determine if imaging
of oocytes can be developed as a non-invasive method for the
selection of oocytes with greater developmental competence.
Briefly, it is important to mention that light exposure to
oocytes and embryos can be detrimental to subsequent
development [186] and must be considered in all microscopy
evaluations (review see: [187]).
Polarized light microscopy
The use of polarized optics to evaluate gametes and embryos
is not a new idea and has been extensively reviewed else-
where [20]. Polarized light is a contrast-enhancing method
that yields quality images obtained by birefringent materials
that have structured organization. A goal of this, and other
imaging technologies discussed below, is non-invasive imaging
of intracellular organelle, structures, and/or processes that
provided informative guidance of selection or developmental
competence. In oocytes, polarized optics can allow visual-
ization of the spindle [188] and organization of the oocyte-
surrounding ZP [189]. One of the advantages of polarized
light microscopy is that it can be performed in real time and
on living cells. Clinical use of polarized optics with human
oocytes demonstrated that the metaphase II MS is not always
directly adjacent or beneath the extruded PBI [190]. This is
of potential interest, especially in oocytes used for ICSI, when
the alignment of the injection plane is based on the position
of the oocyte polar body. This observation also stimulates the
query of why and how does the extrusion of the first meiotic
polar body not align with the subsequent meiotic metaphase
spindle? This may likely be due to manual manipulation of
the oocyte with pipets during the removal of cumulus cells
in denuding manipulations—resulting in displacement of the
polar body in the PVS.
There have been studies focused on the use of polarized
optics to detect the presence or absence of the oocyte MS and
subsequent implantation and pregnancy rates. Unfortunately,
these studies have contradictory results [24,25]—indicating
that this live-cell imaging of the oocyte MS still warrants well-
controlled evaluation. These types of studies, as well as the
sensitivity of the oocyte MS to temperature as visualized with
polarized microscopy [191], emphasizes that the MS and the
tubulin polymerization is a dynamic structure/process. They
also stress the importance of considering time and space in
the dynamic evaluation of meiosis and the spindle. De Santis
and coworkers attempted to use spindle imaging via polarized
light microscopy as a predictor of oocyte quality [108], but
concluded that while there was a trend observed between
low oocyte spindle retardance and poor subsequent embryo
quality, the association was not statistically significant. What
may have been the most important report in this manuscript
was the observation that many oocytes that have extruded
the PBIhad not actually completed chromatin segregation
and were actually still in telophase I when one was able to
visualize the spindle. This observation could have significant
importance in considering the timing of sperm injection in
ICSI. If our light microscopy observations of the presence
of a polar body are not always indicative of completion of
meiosis I, progression to MII, and chromatic segregation,
then the injection process may be causing unrecognizable
damage to the telophase I spindle in the process of segregating
chromatin. This could be a future utility of polarized light
microscopy in non-invasive assessment of human oocytes with
a beneficial impact on subsequent normal fertilization and
euploid embryo development.
Raman microspectroscopy
This is a combination of Raman spectroscopy and confocal
microscopy and is founded on principles of inelastic scatter-
ing, which yield interactions of light and matter. The photon
scattering results in unique spectra that can be used to identify
molecules and their molecular bonds in living cells. We are
specifically discussing this technology over other technologies
such as multiphoton excitation fluorescence microscopy [192]
and light-sheet fluorescence microscopy [193], because of the
lack of labeled probes necessary for excitation, observation,
quantification. With that said, it is important to recognize that
these types of imaging systems (multiphoton and light-sheet)
are continuously developing and comparatively have distin-
guished themselves for having very low phototoxicity and
high spatial and temporal resolution—making them ideal for
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
10 Non-invasive oocyte quality assessment
Figure 2. Composite schematic, micrographs, and graphic quantification of oocyte lipids with coherent anti-Stokes Raman scattering (CARS)
microscopy. (A) Schematic of CARS design. A single laser source provides a narrowband pump pulse (green) as well as a broadband Stokes pulse (red)
using a photonic crystal fiber (PCF). With a broadband source, two methods of detection were used for live-cell imaging: univariate photomultiplier tube
(PMT; requires less time to capture images) and multivariate charged-coupled device (CCD) CARS imaging. FI: Faraday Isolator, GP: Glan-Taylor Prism,
BP/LP/SP: band/long/short pass filter, DM: dichroic mirror, PCF: photonic crystal fiber. (B) Lipid content in mouse oocyte during meiosis as assessed and
quantified with CARS-PMT demonstrated that oocytes have low lipid content when they are GV-I meiotically incompetent (MeI). Oocytes gain lipids as
they grow and gain meiotic competence (still GV-I; MeC). As oocytes resume meiosis and progress from GV-I to metaphases II (MII), they lose
cytoplasmic lipids. (C) Oocytes from wild type and genetically obese (ob/ob) were assessed for lipid content with CARS-PMT. In meiotically competent
GV-I oocytes, oocyte lipid content was correlated with body mass. (D) Comparison of non-invasive lipid quantification with live-cell CARS-PMT to
invasive lipid quantification with oocyte fixation and staining with Nile Red. Micrographs represent both CARS-PMT (2850 cm1) and NILE Red
fluorescence with their respective thresholded images for oocytes from human, mouse, cow, and pig. Graphically represented is quantification, showing
good agreement between CARS and fluorescence and expected higher intra-oocyte lipid in pig and cow compared to mouse and human. Scale bar is
50 μm. Figure adapted from [199] with permission from the Analyst, The Royal Society of Chemistry .
studies of development and cellular dynamics [194]. Raman
microspectroscopy has been used to evaluate mouse oocytes
[195]. These researchers found that using both synchrotron
Fourier transform-infrared and Raman microspectroscopy,
they were able to identify patterns of intracellular lipids and
areas of high protein content (assumed to be mitochondria)
that were different as oocytes matured. Such reports hold
promise as proof of concepts and require further investigation
to determine if Raman microspectroscopy will yield informa-
tive data in selecting oocytes beyond maturity, which can in
and of itself be assessed by standard light microcopy. Xenopus
oocytes have been evaluated using Raman microspectroscopy
to characterize the presence, absence, and cytoplasmic dis-
tribution of carotenoids [196]. An important component of
this investigation was the reported lack of detrimental impact
of Raman microscopy on Xenopus oocyte viability. Finally,
in a quite relevant report, Bogliolo and colleagues investi-
gated mouse aging and associated oxidative damage of MII
oocytes with Raman imaging. Using principal component
analysis, they showed distinguishable spectra between oocytes
from young mice versus those obtained from in vitro-aged,
oxidative-damaged, and old oocytes—specifically, significant
differences in lipid and protein components [197]. If this
held true for human oocytes, it could be an important non-
invasive means of oocyte assessment and selection. With
that said, although Raman spectroscopy is considered non-
invasive, the long imaging times required with spontaneous
Raman tend to counterindicate its use for live-cell imaging of
human gametes and embryos, thus potentially limiting its use
in clinical ART. Finally, Raman microspectroscopy has also
been used to evaluate oocyte responses to technical exposures,
such as vitrification and warming [198]. The sheep oocytes’
ZP carbohydrate secondary structures were altered by the
event of vitrification and warming, but not by cryoprotectant
exposure alone. These are interesting applications of Raman
imaging to address practical knowledge gaps at a molecu-
lar level. One would not be surprised to see future studies
using Raman imaging to gain non-invasive information about
oocyte selection.
A slight permutation of Raman microspectroscopy is
the use of coherent anti-Stokes Raman scattering or CARS
microscopy. Coherent anti-Stokes Raman scattering micros-
copy is considered a nonlinear vibrational microscopy
that has recently been demonstrated to allow non-invasive
quantification of oocyte lipids [199]. In relation to the
non-invasive nature of CARS, studies were performed to
assess CARS laser exposure (2–3 min) of mouse zygotes and
subsequent embryo development. As mentioned previously,
these types of experiments are essential to determine if the
technology is truly non-invasive and of potential clinical
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 11
relevance. It was found that the CARS laser exposure that is
compatible with intra-oocyte lipid quantification did not have
any detrimental effect on subsequent blastocyst development
rates (n=52 zygotes; 85%) compared to sham CARS laser
exposure controls (n=48 zygotes; 83%). Granted, this is
only a first step in determining the non-invasive influence of
CARS microscopy on embryo development in the mouse; it
also needs to be determined with human oocytes. Experiments
went on to demonstrate that the use of a broadband excitation
source allowed for single-wavelength images collected by
way of a photomultiplier tube (CARS-PMT) and could also
support the acquisition of the entire vibrational spectrum
for each pixel by way of charge-coupled device (CARS-
CCD). The CARS-PMT was shown to allow quantification
of intra-oocyte lipids similar to CARS-CCD (and Nile Red
staining as a control; invasive terminal experimental arm
where oocytes require fixation and staining); yet the CARS-
PMT required less time and laser exposure. Experiments then
demonstrated that intra-oocyte lipid quantification could be
performed on porcine, bovine, murine, and human oocytes
with CARS-PMT yielding expected species differences in
lipid droplet quantities (Figure 2A). Of interest was that
human oocytes assessed contained very few lipid droplets
in comparison to other species. What this might mean in
relation to human oocytes, energy stores in the form of
lipid droplets, and subsequent embryonic developmental
competence was discussed [199]. It was also demonstrated
that CARS-PMT could quantify statistically different levels of
lipid droplets in mouse oocytes of different meiotic stages
(growing meiotically incompetent germinal vesicle-intact
versus fully grown meiotically competent germinal vesicle-
intact versus metaphase II oocytes; Figure 2B). Finally, it was
also demonstrated that mice with different body composi-
tions (obese versus lean) had identifiable and quantifiable
differences in intra-oocyte lipid droplets (Figure 2C). While
these were first in field experiments that require repeating
and additional investigative queries to determine clinical
application, they are examples of the power of integrating
novel microscopic systems to gamete/embryo biological and
potentially practical studies. The authors concluded that
CARS-PMT appears to be a non-invasive live-cell imaging
technique of lipid quantification that (1) has experimental
power in basic cell biology, (2) may have practical utility
for identifying developmental predictive biomarkers while
advancing biology-based oocyte/embryo selection, and (3)
may have the ability to yield rational supporting technology
for decision-making in rodents, domestic species, and
human-assisted reproduction and/or fertility preservation.
Future experiments will examine the application of CARS
microscopy, as well as other novel means of viewing the intra-
structure, organization, and function of oocytes, to glean
biomarker data on oocyte developmental competences and
selection/deselection of oocytes in ARTs.
Conclusions and future directions in
non-invasive oocyte quality assessments
The integration of chemistry, genomics, microfluidics,
microscopy, physics, and other biomedical engineering
technologies into the basic studies of oocyte biology, and in
testing and perfecting practical solution of oocyte evaluation,
are the future for non-invasive assessment of oocytes.
Recent investigative and practical platforms for single oocyte
evaluations are in development (Figure 2)[200]. These will
likely be applied in the evaluation of oocyte transcriptomics
[201], single-cell polar body genomics [202], mitochondrial
genomics [203], metabolomics [180], and potentially non-
invasive genetic analysis of oocyte spent culture media similar
to current evaluations of non-invasive preimplantation genetic
testing for aneuploidy of embryos [204,205]. These goals are
not easy and have yet to be completely brought to fruition,
but progress is being made. It will be essential to foster and
maintain cross-discipline investigation in order to continue
building foundational, rigorous science that will determine
what represents a reliable biomarker of oocyte quality and
developmental competence, while in tandem developing
technologies and platforms that are truly non-invasive and
compatible with clinical laboratory utility in the realm of
ARTs. Basic and applied science are equally important in this
journey toward identification and application of non-invasive
oocyte assessment.
Acknowledgments
We would like to thank Mrs. Sarah Block for editorial comments. We
appreciate the content review and critiques by Dra. Martha Isolina
Garcia Amador. We acknowledge the American Society of Reproductive
Medicine Research Institute funding for partial support of manuscript
production. Finally, we would also like to acknowledge the scientific
contributions of many researchers in oocyte biology and assessment that
unfortunately have not been referenced in this review due to content and
size constraints.
Author contributions
RS, DM, and GDS conceptualized and wrote this manuscript.
Conflict of Interest
RS and DM have no conflicts of interest. GDS functions as a consultant
to Overture Life, a company focused on microfluidics and automation
of gamete and embryo isolation, manipulation, culture, assessment, and
cryopreservation.
References
1. Gilchrist RB, Lane M, Thompson JG. Oocyte-secreted factors:
regulators of cumulus cell function and oocyte quality. Hum
Reprod Update 2008; 14:159–177.
2. Mtango NR, Potireddy S, Latham KE. Oocyte quality and mater-
nal control of development. Int Rev Cell Mol Biol 2008; 268:
223–290.
3. Humaidan P, Alviggi C, Fischer R, Esteves SC. The novel POSEI-
DON stratification of ’Low prognosis patients in assisted repro-
ductive technology’ and its proposed marker of successful out-
come. F1000Res 2016; 5:2911.
4. Van Soom A, Vandaele L, Goossens K, de Kruif A, Peelman L.
Gamete origin in relation to early embryo development. Theri-
ogenology 2007; 68:S131–S137.
5. Swain JE, Pool TB. ART failure: oocyte contributions to
unsuccessful fertilization. Hum Reprod Update 2008; 14:
431–446.
6. Mahutte NG, Arici A. Failed fertilization: is it predictable? Curr
Opin Obstet Gynecol 2003; 15:211–218.
7. Marteil G, Richard-Parpaillon L, Kubiak JZ. Role of oocyte qual-
ity in meiotic maturation and embryonic development. Reprod
Biol 2009; 9:203–224.
8. Nagy ZP, Jones-Colon S, Roos P, Botros L, Greco E, Dasig J, Behr
B. Metabolomic assessment of oocyte viability. Reprod Biomed
Online 2009; 18:219–225.
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
12 Non-invasive oocyte quality assessment
9. Patrizio P, Fragouli E, Bianchi V, Borini A, Wells D. Molecular
methods for selection of the ideal oocyte. Reprod Biomed Online
2007; 15:346–353.
10. Combelles CM, Racowsky C. Assessment and optimization of
oocyte quality during assisted reproductive technology treatment.
Semin Reprod Med 2005; 23:277–284.
11. Centers for Disease Control and Prevention. American Society
for Reproductive Medicine, Society for Assisted Reproductive
Technology 2016 Assisted Reproductive Technology National
Summary Report. Atlanta, GA: U.S. Department of Health and
Human Services; 2018.
12. Forman EJ, Hong KH, Ferry KM, Tao X, Taylor D, Levy B,
Treff NR, Scott RT Jr. In vitro fertilization with single euploid
blastocyst transfer: a randomized controlled trial. Fertil Steril
2013; 100:100, e101–107.
13. Rienzi L, Vajta G, Ubaldi F. Predictive value of oocyte morphol-
ogy in human IVF: a systematic review of the literature. Hum
Reprod Update 2011; 17:34–45.
14. Tiitinen A. Single embryo transfer: why and how to identify the
embryo with the best developmental potential. Best Pract Res Clin
Endocrinol Metab 2019; 33:77–88.
15. Ciepiela P, Baczkowski T, Drozd A, Kazienko A, Stachowska
E, Kurzawa R. Arachidonic and linoleic acid derivatives impact
oocyte ICSI fertilization—a prospective analysis of follicular fluid
and a matched oocyte in a ’one follicle—one retrieved oocyte—
one resulting embryo’ investigational setting. PLoS One 2015;
10:e0119087.
16. Millbank J, Stuhmcke A, Karpin I. Embryo donation and under-
standing of kinship: the impact of law and policy. Hum Reprod
2017; 32:133–138.
17. Kort J, Behr B. Biomechanics and developmental potential of
oocytes and embryos. Fertil Steril 2017; 108:738–741.
18. Rienzi L, Ubaldi F, Martinez F, Iacobelli M, Minasi MG, Fer-
rero S, Tesarik J, Greco E. Relationship between meiotic spindle
location with regard to the polar body position and oocyte
developmental potential after ICSI. Hum Reprod 2003; 18:
1289–1293.
19. Montag M, Schimming T, Koster M, Zhou C, Dorn C, Rosing
B, van der Ven H, Ven der Ven K. Oocyte zona birefringence
intensity is associated with embryonic implantation potential in
ICSI cycles. Reprod Biomed Online 2008; 16:239–244.
20. Montag M, Koster M, van der Ven K, van der Ven H. Gamete
competence assessment by polarizing optics in assisted reproduc-
tion. Hum Reprod Update 2011; 17:654–666.
21. Wang WH, Meng L, Hackett RJ, Odenbourg R, Keefe DL. The
spindle observation and its relationship with fertilization after
intracytoplasmic sperm injection in living human oocytes. Fertil
Steril 2001; 75:348–353.
22. Shen Y, Stalf T, Mehnert C, De Santis L, Cino I, Tinneberg HR,
Eichenlaub-Ritter U. Light retardance by human oocyte spindle is
positively related to pronuclear score after ICSI. Reprod Biomed
Online 2006; 12:737–751.
23. Rienzi L, Ubaldi F, Iacobelli M, Minasi MG, Romano S, Greco
E. Meiotic spindle visualization in living human oocytes. Reprod
Biomed Online 2005; 10:192–198.
24. Madaschi C, de Souza Bonetti TC, de Almeida Ferreira Braga DP,
Pasqualotto FF, Iaconelli A Jr, Borges E Jr. Spindle imaging: a
marker for embryo development and implantation. Fertil Steril
2008; 90:194–198.
25. Chamayou S, Ragolia C, Alecci C, Storaci G, Maglia E, Russo E,
Guglielmino A. Meiotic spindle presence and oocyte morphology
do not predict clinical ICSI outcomes: a study of 967 transferred
embryos. Reprod Biomed Online 2006; 13:661–667.
26. Fang C, Tang M, Li T, Peng WL, Zhou CQ, Zhuang GL, Leong
M. Visualization of meiotic spindle and subsequent embryonic
development in in vitro and in vivo matured human oocytes. J
Assist Reprod Genet 2007; 24:547–551.
27. Moon JH, Hyun CS, Lee SW, Son WY, Yoon SH, Lim JH.
Visualization of the metaphase II meiotic spindle in living human
oocytes using the Polscope enables the prediction of embryonic
developmental competence after ICSI. Hum Reprod 2003; 18:
817–820.
28. Mogessie B, Scheffler K, Schuh M. Assembly and positioning of
the oocyte meiotic spindle. Annu Rev Cell Dev Biol 2018; 34:
381–403.
29. Taylor TH, Chang CC, Elliott T, Colturato LF, Kort HI, Nagy
ZP. Effect of denuding on polar body position in in-vitro matured
oocytes. Reprod Biomed Online 2008; 17:515–519.
30. Rienzi L, Ubaldi F. Oocyte retrieval and selection. In: Gardner D,
Weissman A, Howles C, Shoham Z (eds.), Textbook of Assisted
Reproductive Technologies: Laboratory and Clinical Perspectives,
3rd ed. London, UK: Informa Healthcare; 2009: 5–101.
31. Mihajlovic AI, FitzHarris G. Segregating chromosomes in the
mammalian oocyte. Curr Biol 2018; 28:R895–R907.
32. He M, Zhang T, Yang Y, Wang C. Mechanisms of oocyte matura-
tion and related epigenetic regulation. Front Cell Dev Biol 2021;
9:654028.
33. Trebichalska Z, Kyjovska D, Kloudova S, Otevrel P, Hampl A,
Holubcova Z. Cytoplasmic maturation in human oocytes: an
ultrastructural study dagger. Biol Reprod 2021; 104:106–116.
34. Mehta P, Pauly M. Single vs multiple embryo transfer: com-
parative costs and a call for change. JAMA Pediatr 2014; 168:
997–998.
35. De Sutter P, Dozortsev D, Qian C, Dhont M. Oocyte morphology
does not correlate with fertilization rate and embryo quality
after intracytoplasmic sperm injection. Hum Reprod 1996; 11:
595–597.
36. Ten J, Mendiola J, Vioque J, de Juan J, Bernabeu R. Donor oocyte
dysmorphisms and their influence on fertilization and embryo
quality. Reprod Biomed Online 2007; 14:40–48.
37. Rienzi L, Ubaldi FM, Iacobelli M, Minasi MG, Romano S, Ferrero
S, Sapienza F, Baroni E, Litwicka K, Greco E. Significance of
metaphase II human oocyte morphology on ICSI outcome. Fertil
Steril 2008; 90:1692–1700.
38. Esfandiari N, Burjaq H, Gotlieb L, Casper RF. Brown oocytes:
implications for assisted reproductive technology. Fertil Steril
2006; 86:1522–1525.
39. Balaban B, Ata B, Isiklar A, Yakin K, Urman B. Severe cytoplasmic
abnormalities of the oocyte decrease cryosurvival and subsequent
embryonic development of cryopreserved embryos. Hum Reprod
2008; 23:1778–1785.
40. Loutradis D, Drakakis P, Kallianidis K, Milingos S, Dendrinos S,
Michalas S. Oocyte morphology correlates with embryo quality
and pregnancy rate after intracytoplasmic sperm injection. Fertil
Steril 1999; 72:240–244.
41. Wilding M, Di Matteo L, D’Andretti S, Montanaro N, Capo-
bianco C, Dale B. An oocyte score for use in assisted reproduction.
J Assist Reprod Genet 2007; 24:350–358.
42. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elas-
ticity directs stem cell lineage specification. Cell 2006; 126:
677–689.
43. Ebner T, Moser M, Sommergruber M, Puchner M, Wiesinger R,
Tews G. Developmental competence of oocytes showing increased
cytoplasmic viscosity. Hum Reprod 2003; 18:1294–1298.
44. Ebner T, Balaban B, Moser M, Shebl O, Urman B, Ata B, Tews G.
Automatic user-independent zona pellucida imaging at the oocyte
stage allows for the prediction of preimplantation development.
Fertil Steril 2010; 94:913–920.
45. Dumoulin JM, Coonen E, Bras M, Bergers-Janssen JM, Ignoul-
Vanvuchelen RC, van Wissen LC, Geraedts JP, Evers JL. Embryo
development and chromosomal anomalies after ICSI: effect of the
injection procedure. Hum Reprod 2001; 16:306–312.
46. Yanez LZ, Han J, Behr BB, Pera RAR, Camarillo DB. Human
oocyte developmental potential is predicted by mechanical prop-
erties within hours after fertilization. Nat Commun 2016;
7:10809.
47. Fortune JE. Ovarian follicular growth and development in mam-
mals. Biol Reprod 1994; 50:225–232.
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 13
48. Revelli A, Delle Piane L,Casano S, Molinari E, Massobrio M, Rin-
audo P. Follicular fluid content and oocyte quality: from single
biochemical markers to metabolomics. Reprod Biol Endocrinol
2009; 7:40.
49. Assou S, Anahory T, Pantesco V, Le Carrour T, Pellestor F, Klein
B, Reyftmann L, Dechaud H, De Vos J, Hamamah S. The human
cumulus–oocyte complex gene-expression profile. Hum Reprod
2006; 21:1705–1719.
50. Kocabas AM, Crosby J, Ross PJ, Otu HH, Beyhan Z, Can H, Tam
WL, Rosa GJ, Halgren RG, Lim B, Fernandez E, Cibelli JB. The
transcriptome of human oocytes. Proc Natl Acad Sci U S A 2006;
103:14027–14032.
51. Jiao J, Shi B, Wang T, Fang Y, Cao T, Zhou Y, Wang X, Li D.
Characterization of long non-coding RNA and messenger RNA
profiles in follicular fluid from mature and immature ovarian
follicles of healthy women and women with polycystic ovary
syndrome. Hum Reprod 2018; 33:1735–1748.
52. Zamah AM, Hassis ME, Albertolle ME, Williams KE. Proteomic
analysis of human follicular fluid from fertile women. Clin Pro-
teomics 2015; 12:5.
53. Kim YS, Kim MS, Lee SH, Choi BC, Lim JM, Cha KY, Baek
KH. Proteomic analysis of recurrent spontaneous abortion: iden-
tification of an inadequately expressed set of proteins in human
follicular fluid. Proteomics 2006; 6:3445–3454.
54. 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 preim-
plantation embryo: a randomized controlled trial. Hum Reprod
2007; 22:980–988.
55. Delhanty JD, Penketh RJ. Cytogenetic analysis of unfertilized
oocytes retrieved after treatment with the LHRH analogue,
buserelin. Hum Reprod 1990; 5:699–702.
56. Elbling L, Colot M. Abnormal development and transport and
increased sister-chromatid exchange in preimplantation embryos
following superovulation in mice. Mutat Res 1985; 147:189–195.
57. Magli MC, Ferraretti AP, Crippa A, Lappi M, Feliciani E, Gia-
naroli L. First meiosis errors in immature oocytes generated by
stimulated cycles. Fertil Steril 2006; 86:629–635.
58. Massie JA, Shahine LK, Milki AA, Westphal LM, Lathi RB.
Ovarian stimulation and the risk of aneuploid conceptions. Fertil
Steril 2011; 95:970–972.
59. Munne S, Magli C, Adler A, Wright G, de Boer K, Mortimer
D, Tucker M, Cohen J, Gianaroli L. Treatment-related chromo-
some abnormalities in human embryos. Hum Reprod 1997; 12:
780–784.
60. Verpoest W, Fauser BC, Papanikolaou E, Staessen C, Van Landuyt
L, Donoso P, Tournaye H, Liebaers I, Devroey P. Chromosomal
aneuploidy in embryos conceived with unstimulated cycle IVF.
Hum Reprod 2008; 23:2369–2371.
61. Hong KH, Franasiak JM, Werner MM, Patounakis G, Juneau
CR, Forman EJ, Scott RT Jr. Embryonic aneuploidy rates are
equivalent in natural cycles and gonadotropin-stimulated cycles.
Fertil Steril 2019; 112:670–676.
62. Takahashi C, Fujito A, Kazuka M, Sugiyama R, Ito H, Isaka
K. Anti-Mullerian hormone substance from follicular fluid is
positively associated with success in oocyte fertilization during
in vitro fertilization. Fertil Steril 2008; 89:586–591.
63. O’Brien Y, Wingfield M, O’Shea LC. Anti-Mullerian hormone
and progesterone levels in human follicular fluid are predictors of
embryonic development. Reprod Biol Endocrinol 2019; 17:47.
64. Tesarik J, Mendoza C. Direct non-genomic effects of follicular
steroids on maturing human oocytes: oestrogen versus androgen
antagonism. Hum Reprod Update 1997; 3:95–100.
65. Teissier MP, Chable H, Paulhac S, Aubard Y. Comparison of
follicle steroidogenesis from normal and polycystic ovaries in
women undergoing IVF: relationship between steroid concentra-
tions, follicle size, oocyte quality and fecundability. Hum Reprod
2000; 15:2471–2477.
66. Artini PG, Battaglia C, D’Ambrogio G, Barreca A, Droghini F,
Volpe A, Genazzani AR. Relationship between human oocyte
maturity, fertilization and follicular fluid growth factors. Hum
Reprod 1994; 9:902–906.
67. Reinthaller A, Deutinger J, Bieglmayer C, Riss P, Muller-Tyl E,
Fischl F, Janisch H. Hormonal parameters in follicular fluid and
the fertilization rate of in vitro cultured oocytes. Arch Gynecol
1987; 240:207–210.
68. Kreiner D, Liu HC, Itskovitz J, Veeck L, Rosenwaks Z. Follicular
fluid estradiol and progesterone are markers of preovulatory
oocyte quality. Fertil Steril 1987; 48:991–994.
69. Botero-Ruiz W, Laufer N, DeCherney AH, Polan ML, Haseltine
FP, Behrman HR. The relationship between follicular fluid steroid
concentration and successful fertilization of human oocytes in
vitro. Fertil Steril 1984; 41:820–826.
70. Subramanian MG, Sacco AG, Moghissi KS, Magyar DM, Hayes
MF, Lawson DM, Gala RR. Human follicular fluid: prolactin is
biologically active and ovum fertilization correlates with estra-
diol concentration. J In Vitro Fert Embryo Transf 1988; 5:
129–133.
71. Lee MS, Ben-Rafael Z, Meloni F, Mastroianni L Jr, Flickinger GL.
Relationship of human oocyte maturity, fertilization, and cleavage
to follicular fluid prolactin and steroids. J In Vitro Fert Embryo
Transf 1987; 4:168–172.
72. Tarlatzis BC, Laufer N, DeCherney AH, Polan ML, Haseltine FP,
Behrman HR. Adenosine 3,5-monophosphate levels in human
follicular fluid: relationship to oocyte maturation and achieve-
ment of pregnancy after in vitro fertilization. J Clin Endocrinol
Metab 1985; 60:1111–1115.
73. Tarlatzis BC, Pazaitou K, Bili H, Bontis J, Papadimas J, Lagos
S, Spanos E, Mantalenakis S. Growth hormone, oestradiol, pro-
gesterone and testosterone concentrations in follicular fluid after
ovarian stimulation with various regimes for assisted reproduc-
tion. Hum Reprod 1993; 8:1612–1616.
74. Costa LO, Mendes MC, Ferriani RA, Moura MD, Reis RM, Silva
de Sa MF. Estradiol and testosterone concentrations in follicular
fluid as criteria to discriminate between mature and immature
oocytes. Braz J Med Biol Res 2004; 37:1747–1755.
75. Berger MA, Laufer N, Lewin A, Navot D, Rabinowitz R, Eisen-
berg S, Margalioth EJ, Schenker JG. Cholesterol and steroid levels
in human follicular fluids of human menopausal gonadotropin-
induced cycles for in vitro fertilization. J In Vitro Fert Embryo
Transf 1987; 4:30–33.
76. Rosenbusch B, Djalali M, Sterzik K. Is there any correlation
between follicular f luid hormone concentrations, fertilizability,
and cytogenetic analysis of human oocytes recovered for in vitro
fertilization? Fertil Steril 1992; 57:1358–1360.
77. Messinis IE, Templeton AA. Relationship between intrafollicular
levels of prolactin and sex steroids and in-vitro fertilization of
human oocytes. Hum Reprod 1987; 2:607–609.
78. Suchanek E, Simunic V, Macas E, Kopjar B, Grizelj V.
Prostaglandin F2 alpha, progesterone and estradiol concentra-
tions in human follicular fluid and their relation to success of in
vitro fertilization. Eur J Obstet Gynecol Reprod Biol 1988; 28:
331–339.
79. Mo Q, Lu S-, Simon NG. Dehydroepiandrosterone and its
metabolites: differential effects on androgen receptor trafficking
and transcriptional activity. J Steroid Biochem Mol Biol 2006; 99:
50–58.
80. Bonser J, Walker J, Purohit A, Reed MJ, Potter BV, Willis DS,
Franks S, Mason HD. Human granulosa cells are a site of sul-
phatase activity and are able to utilize dehydroepiandrosterone
sulphate as a precursor for oestradiol production. J Endocrinol
167:465–471.
81. Dehennin L, Jondet M, Scholler R. Androgen and 19-norsteroid
profiles in human preovulatory follicles from stimulated cycles:
an isotope dilution-mass spectrometric study. J Steroid Biochem
1987; 26:399–405.
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
14 Non-invasive oocyte quality assessment
82. Hossein Rashidi B, Hormoz B, Shahrokh Tehraninejad E, Shariat
M, Mahdavi A. Testosterone and dehydroepiandrosterone sul-
phate levels and IVF/ICSI results. Gynecol Endocrinol 2009; 25:
194–198.
83. Li L, Ferin M, Sauer MV, Lobo RA. Dehydroepiandrosterone
in follicular fluid is produced locally, and levels correlate nega-
tively with in vitro fertilization outcomes. Fertil Steril 2011; 95:
1830–1832.
84. Chimote NM, Nath NM, Chimote NN, Chimote BN. Follic-
ular fluid dehydroepiandrosterone sulfate is a credible marker
of oocyte maturity and pregnancy outcome in conventional
in vitro fertilization cycles. J Hum Reprod Sci 2015; 8:
209–213.
85. Smith MP, Flannery GR, Randle BJ, Jenkins JM, Holmes CH.
Leukocyte origin and profile in follicular aspirates at oocyte
retrieval. Hum Reprod 2005; 20:3526–3531.
86. Seli E, Robert C, Sirard MA. OMICS in assisted reproduction:
possibilities and pitfalls. Mol Hum Reprod 2010; 16:513–530.
87. Anderson RA, Sciorio R, Kinnell H, Bayne RA, Thong KJ, de
Sousa PA, Pickering S.Cumulus gene expression as a predictor of
human oocyte fertilisation, embryo development and competence
to establish a pregnancy. Reproduction 2009; 138:629–637.
88. McKenzie LJ, Pangas SA, Carson SA, Kovanci E, Cisneros P,
Buster JE, Amato P, Matzuk MM. Human cumulus granulosa cell
gene expression: a predictor of fertilization and embryo selection
in women undergoing IVF. Hum Reprod 2004; 19:2869–2874.
89. Cillo F, Brevini TA, Antonini S, Paffoni A, Ragni G, Gandolfi F.
Association between human oocyte developmental competence
and expression levels of some cumulus genes. Reproduction 2007;
134:645–650.
90. Wathlet S, Adriaenssens T, Segers I, Verheyen G, Janssens R,
Coucke W, Devroey P, Smitz J. New candidate genes to predict
pregnancy outcome in single embryo transfer cycles when using
cumulus cell gene expression. Fertil Steril 2012; 98:432–439 432–
439.e4.
91. Hamel M, Dufort I, Robert C, Gravel C, Leveille MC, Leader A,
Sirard MA. Identification of differentially expressed markers in
human follicular cells associated with competent oocytes. Hum
Reprod 2008; 23:1118–1127.
92. Hamel M, Dufort I, Robert C, Leveille MC, Leader A, Sirard
MA. Genomic assessment of follicular marker genes as pregnancy
predictors for human IVF. Mol Hum Reprod 2010; 16:87–96.
93. Gebhardt KM, Feil DK, Dunning KR, Lane M, Russell DL.
Human cumulus cell gene expression as a biomarker of pregnancy
outcome after single embryo transfer. Fertil Steril 2011; 96:47,
e42–52.
94. Uyar A, Seli E. Embryo assessment strategies and their validation
for clinical use: a critical analysis of methodology. Curr Opin
Obstet Gynecol 2012; 24:141–150.
95. Shutt DA, Lopata A. The secretion of hormones during the culture
of human preimplantation embryos with corona cells. Fertil Steril
1981; 35:413–416.
96. Tanghe S, Van Soom A, Nauwynck H, Coryn M, de Kruif A.
Minireview: functions of the cumulus oophorus during oocyte
maturation, ovulation, and fertilization. Mol Reprod Dev 2002;
61:414–424.
97. Dekel N, Beers WH. Development of the rat oocyte in vitro:
inhibition and induction of maturation in the presence or absence
of the cumulus oophorus. Dev Biol 1980; 75:247–254.
98. Larsen WJ, Wert SE, Brunner GD. A dramatic loss of cumulus cell
gap junctions is correlated with germinal vesicle breakdown in rat
oocytes. Dev Biol 1986; 113:517–521.
99. Hsieh M, Zamah AM, Conti M. Epidermal growth factor-like
growth factors in the follicular fluid: role in oocyte development
and maturation. Semin Reprod Med 2009; 27:52–61.
100. Elvin JA, Clark AT, Wang P, Wolfman NM, Matzuk MM.
Paracrine actions of growth differentiation factor-9 in the mam-
malian ovary. Mol Endocrinol 1999; 13:1035–1048.
101. Pangas SA, Matzuk MM. The art and artifact of GDF9 activity:
cumulus expansion and the cumulus expansion-enabling factor.
Biol Reprod 2005; 73:582–585.
102. Uyar A, Torrealday S, Seli E. Cumulus and granulosa cell markers
of oocyte and embryo quality. Fertil Steril 2013; 99:979–997.
103. McReynolds S, Dzieciatkowska M, McCallie BR, Mitchell SD,
Stevens J, Hansen K, Schoolcraft WB, Katz-Jaffe MG. Impact of
maternal aging on the molecular signature of human cumulus
cells. Fertil Steril 2012; 98:1574, e1575–1580.
104. Eichenlaub-Ritter U, Schmiady H, Kentenich H, Soewarto D.
Recurrent failure in polar body formation and premature chro-
mosome condensation in oocytes from a human patient: indica-
tors of asynchrony in nuclear and cytoplasmic maturation. Hum
Reprod 1995; 10:2343–2349.
105. Ebner T, Moser M, Yaman C, Feichtinger O, Hartl J, Tews
G. Elective transfer of embryos selected on the basis of first
polar body morphology is associated with increased rates of
implantation and pregnancy. Fertil Steril 1999; 72:599–603.
106. Ebner T, Moser M, Sommergruber M, Yaman C, Pf leger U, Tews
G. First polar body morphology and blastocyst formation rate in
ICSI patients. Hum Reprod 2002; 17:2415–2418.
107. Fancsovits P, Tothne ZG, Murber A, Takacs FZ, Papp Z, Urbanc-
sek J. Correlation between first polar body morphology and
further embryo development. Acta Biol Hung 2006; 57:331–338.
108. De Santis L, Cino I, Rabellotti E, Calzi F, Persico P, Borini A,
Coticchio G. Polar body morphology and spindle imaging as
predictors of oocyte quality. Reprod Biomed Online 2005; 11:
36–42.
109. Verlhac MH, Lefebvre C, Guillaud P, Rassinier P, Maro B. Asym-
metric division in mouse oocytes: with or without Mos. Curr Biol
2000; 10:1303–1306.
110. Gorbsky GJ. The spindle checkpoint and chromosome segrega-
tion in meiosis. FEBS J 2015; 282:2471–2487.
111. Nagaoka SI, Hassold TJ, Hunt PA. Human aneuploidy: mecha-
nisms and new insights into an age-old problem. Nat Rev Genet
2012; 13:493–504.
112. Giudice LC. Environmental toxicants: hidden players on the
reproductive stage. Fertil Steril 2016; 106:791–794.
113. Handyside AH, Kontogianni EH, Hardy K, Winston RM. Preg-
nancies from biopsied human preimplantation embryos sexed by
Y-specific DNA amplification. Nature 1990; 344:768–770.
114. Verlinsky Y, Cieslak J, Ivakhnenko V, Evsikov S, Wolf G, White
M, Lifchez A, Kaplan B, Moise J, Valle J, Ginsberg N, Strom C
et al. Preimplantation diagnosis of common aneuploidies by the
first- and second-polar body FISH analysis. J Assist Reprod Genet
1998; 15:285–289.
115. Verpoest W, Staessen C, Bossuyt PM, Goossens V, Altarescu G,
Bonduelle M, Devesa M, Eldar-Geva T, Gianaroli L, Griesinger G,
Kakourou G, Kokkali G et al. Preimplantation genetic testing for
aneuploidy by microarray analysis of polar bodies in advanced
maternal age: a randomized clinical trial. Hum Reprod 2018; 33:
1767–1776.
116. Spits C, Le Caignec C,De Rycke M, Van Haute L, Van Steirteghem
A, Liebaers I, Sermon K. Optimization and evaluation of single-
cell whole-genome multiple displacement amplification. Hum
Mutat 2006; 27:496–503.
117. Wells D, Sherlock JK, Handyside AH, Delhanty JD. Detailed chro-
mosomal and molecular genetic analysis of single cells by whole
genome amplification and comparative genomic hybridisation.
Nucleic Acids Res 1999; 27:1214–1218.
118. Wells D, Alfarawati S, Fragouli E. Use of comprehensive chromo-
somal screening for embryo assessment: microarrays and CGH.
Mol Hum Reprod 2008; 14:703–710.
119. Harper JC, Harton G. The use of arrays in preimplantation
genetic diagnosis and screening. Fertil Steril 2010; 94:1173–1177.
120. Treff NR, Tao X, Ferry KM, Su J, Taylor D, Scott RT Jr.
Development and validation of an accurate quantitative real-
time polymerase chain reaction-based assay for human blastocyst
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 15
comprehensive chromosomal aneuploidy screening. Fertil Steril
2012; 97:819–824.
121. Fiorentino F, Bono S, Biricik A,Nuccitelli A, Cotroneo E,Cottone
G, Kokocinski F, Michel CE, Minasi MG, Greco E. Application of
next-generation sequencing technology for comprehensive aneu-
ploidy screening of blastocysts in clinical preimplantation genetic
screening cycles. Hum Reprod 2014; 29:2802–2813.
122. Huang J, Yan L, Lu S, Zhao N, Xie XS, Qiao J. Validation of a
next-generation sequencing-based protocol for 24-chromosome
aneuploidy screening of blastocysts. Fertil Steril 2016; 105:
1532–1536.
123. Tortoriello DV, Dayal M, Beyhan Z, Yakut T, Keskintepe L.
Reanalysis of human blastocysts with different molecular genetic
screening platforms reveals significant discordance in ploidy sta-
tus. J Assist Reprod Genet 2016; 33:1467–1471.
124. Kushnir VA, Darmon SK, Albertini DF, Barad DH, Gleicher N.
Effectiveness of in vitro fertilization with preimplantation genetic
screening: a reanalysis of United States assisted reproductive
technology data 2011-2012. Fertil Steril 2016; 106:75–79.
125. Dondorp W, de Wert G. Innovative reproductive technologies:
risks and responsibilities. Hum Reprod 2011; 26:1604–1608.
126. Xu W, Mezencev R, Kim B, Wang L, McDonald J, Sulchek T.
Cell stiffness is a biomarker of the metastatic potential of ovarian
cancer cells. PLoS One 2012; 7:e46609.
127. Evans EA, Hochmuth RM. Membrane viscoelasticity. Biophys J
1976; 16:1–11.
128. Dimitrov DS. Electric field-induced breakdown of lipid bilayers
and cell membranes: a thin viscoelastic film model. J Membr Biol
1984; 78:53–60.
129. Berteau JP, Oyen M, Shefelbine SJ. Permeability and shear mod-
ulus of articular cartilage in growing mice. Biomech Model
Mechanobiol 2016; 15:205–212.
130. Karcher H, Lammerding J, Huang H, Lee RT, Kamm RD,
Kaazempur-Mofrad MR. A three-dimensional viscoelastic model
for cell deformation with experimental verification. Biophys J
2003; 85:3336–3349.
131. Sato M, Ohshima N, Nerem RM. Viscoelastic properties of
cultured porcine aortic endothelial cells exposed to shear stress. J
Biomech 1996; 29:461–467.
132. Shoji Y, Hamaguchi MS, Hiramoto Y. Mechanical properties of
the endoplasm in starfish oocytes. Exp Cell Res 1978; 117:79–87.
133. Kole TP, Tseng Y, Huang L, Katz JL, Wirtz D. Rho kinase
regulates the intracellular micromechanical response of adherent
cells to rho activation. Mol Biol Cell 2004; 15:3475–3484.
134. Bell S, Terentjev EM. Focal adhesion kinase: the reversible molec-
ular Mechanosensor. Biophys J 2017; 112:2439–2450.
135. Coulombe PA, Omary MB. ’Hard’ and ’soft’ principles defining
the structure, function and regulation of keratin intermediate
filaments. Curr Opin Cell Biol 2002; 14:110–122.
136. Strnad P, Windoffer R, Leube RE. Induction of rapid and
reversible cytokeratin filament network remodeling by inhibition
of tyrosine phosphatases. J Cell Sci 2002; 115:4133–4148.
137. Stuyvers BD, Miura M, ter Keurs HE. Dynamics of viscoelastic
properties of rat cardiac sarcomeres during the diastolic interval:
involvement of Ca2+.JPhysiol1997; 502:661–677.
138. Rankin T, Soyal S, Dean J. The mouse zona pellucida: follicu-
logenesis, fertility and pre-implantation development. Mol Cell
Endocrinol 2000; 163:21–25.
139. Wassarman PM.Zona pellucida glycoproteins. JBiolChem2008;
283:24285–24289.
140. Bleil J, Wassarman P. Structure and function of the zona pellucida:
identification and characterization of the proteins of the mouse
oocyte’s zona pellucida. Dev Biol 1980; 76:185–202.
141. Lefievre L, Conner SJ, Salpekar A, Olufowobi O, Ashton P,
Pavlovic B, Lenton W, Afnan M, Brewis IA, Monk M, Hughes
DC, Barratt CL. Four zona pellucida glycoproteins are expressed
in the human. Hum Reprod 2004; 19:1580–1586.
142. Gupta SK, Bhandari B, Shrestha A, Biswal BK, Palaniappan C,
Malhotra SS, Gupta N. Mammalian zona pellucida glycoproteins:
structure and function during fertilization. Cell Tissue Res 2012;
349:665–678.
143. Hartmann JF, Gwatkin RB, Hutchison CF. Early contact inter-
actions between mammalian gametes in vitro: evidence that the
vitellus influences adherence between sperm and zona pellucida.
Proc Natl Acad Sci U S A 1972; 69:2767–2769.
144. Ganguly A, Bansal P, Gupta T, Gupta SK. ’ZP domain’ of human
zona pellucida glycoprotein-1 binds to human spermatozoa and
induces acrosomal exocytosis. Reprod Biol Endocrinol 2010;
8:110.
145. Wolf D. The mammalian egg’s block to polyspermy. In: Mas-
troianni JI, Biggers JD (eds.), Fertilization and Embryonic Devel-
opment in vitro. Springer, Boston, MA. 1981: 183–197.
146. Wassarman PM. Mammalian fertilization: molecular aspects of
gamete adhesion, exocytosis, and fusion. Cell 1999; 96:175–183.
147. Papi M, Brunelli R, Sylla L, Parasassi T, Monaci M, Maulucci
G, Missori M, Arcovito G, Ursini F, De Spirito M. Mechanical
properties of zona pellucida hardening. Eur Biophys J 2010; 39:
987–992.
148. Bedford JM, Cross NL. Normal penetration of rabbit sperma-
tozoa through a trypsin- and acrosin-resistant zona pellucida. J
Reprod Fertil 1978; 54:385–392.
149. Drobnis EZ, Andrew JB, Katz DF. Biophysical properties of the
zona pellucida measured by capillary suction: is zona hardening
a mechanical phenomenon? J Exp Zool 1988; 245:206–219.
150. Cohen J, Inge KL, Suzman M, Wiker SR, Wright G. Videocine-
matography of fresh and cryopreserved embryos: a retrospective
analysis of embryonic morphology and implantation. Fertil Steril
1989; 51:820–827.
151. Palmstierna M, Murkes D, Csemiczky G, Andersson O, Wramsby
H. Zona pellucida thickness variation and occurrence of visible
mononucleated blastomers in preembryos are associated with a
high pregnancy rate in IVF treatment. J Assist Reprod Genet
1998; 15:70–75.
152. Shen Y, Stalf T, Mehnert C, Eichenlaub-Ritter U, Tinneberg
HR. High magnitude of light retardation by the zona pellucida
is associated with conception cycles. Hum Reprod 2005; 20:
1596–1606.
153. Lai D, Takayama S, Smith GD. Recent microf luidic devices for
studying gamete and embryo biomechanics. JBiomech2015; 48:
1671–1678.
154. Yanez LZ, Camarillo DB. Microfluidic analysis of oocyte and
embryo biomechanical properties to improve outcomes in assisted
reproductive technologies. Mol Hum Reprod 2017; 23:235–247.
155. Nicholson JK, Lindon JC, Holmes E. ’Metabonomics’: under-
standing the metabolic responses of living systems to pathophys-
iological stimuli via multivariate statistical analysis of biological
NMR spectroscopic data. Xenobiotica 1999; 29:1181–1189.
156. Singh R, Sinclair KD. Metabolomics: approaches to assess-
ing oocyte and embryo quality. Theriogenology 2007; 68:
S56–S62.
157. Lindon JC, Holmes E, Nicholson JK. So what’s the deal with
metabonomics? Anal Chem 2003; 75:384A–391A.
158. Riekeberg E, Powers R. New frontiers in metabolomics: from
measurement to insight. F1000Res 2017; 6:1148.
159. Steuer AE, Brockbals L, Kraemer T. Metabolomic strategies in
biomarker research-new approach for indirect identification of
drug consumption and sample manipulation in clinical and foren-
sic toxicology? Front Chem 2019; 7:319.
160. Villas-Boas SG, Mas S, Akesson M, Smedsgaard J, Nielsen J. Mass
spectrometry in metabolome analysis. Mass Spectrom Rev 2005;
24:613–646.
161. Thompson JG. The impact of nutrition of the cumulus oocyte
complex and embryo on subsequent development in ruminants. J
Reprod Dev 2006; 52:169–175.
162. Zeron Y, Ocheretny A, Kedar O, Borochov A, Sklan D, Arav
A. Seasonal changes in bovine fertility: relation to developmen-
tal competence of oocytes, membrane properties and fatty acid
composition of follicles. Reproduction 2001; 121:447–454.
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
16 Non-invasive oocyte quality assessment
163. Booth PJ, Humpherson PG, Watson TJ, Leese HJ. Amino
acid depletion and appearance during porcine preimplantation
embryo development in vitro. Reproduction 2005; 130:655–668.
164. Sinclair KD, Rooke JA, McEvoy TG. Regulation of nutrient
uptake and metabolism in pre-elongation ruminant embryos.
Reprod Suppl 2003; 61:371–385.
165. Uhde K, van Tol HTA, Stout TAE, Roelen BAJ. Metabolomic
profiles of bovine cumulus cells and cumulus-oocyte-complex-
conditioned medium during maturation in vitro. Sci Rep 2018;
8:9477.
166. Seli E, Sakkas D, Scott R, Kwok SC, Rosendahl SM, Burns
DH. Noninvasive metabolomic profiling of embryo culture media
using Raman and near-infrared spectroscopy correlates with
reproductive potential of embryos in women undergoing in vitro
fertilization. Fertil Steril 2007; 88:1350–1357.
167. Seli E, Botros L, Sakkas D, Burns DH. Noninvasive metabolomic
profiling of embryo culture media using proton nuclear magnetic
resonance correlates with reproductive potential of embryos in
women undergoing in vitro fertilization. Fertil Steril 2008; 90:
2183–2189.
168. Scott R, Seli E, Miller K, Sakkas D, Scott K, Burns DH. Noninva-
sive metabolomic profiling of human embryo culture media using
Raman spectroscopy predicts embryonic reproductive potential:
a prospective blinded pilot study. Fertil Steril 2008; 90:77–83.
169. Marhuenda-Egea FC, Martinez-Sabater E, Gonsalvez-Alvarez R,
Lledo B, Ten J, Bernabeu R. A crucial step in assisted reproduction
technology: human embryo selection using metabolomic evalua-
tion. Fertil Steril 2010; 94:772–774.
170. Ahlstrom A, Wikland M, Rogberg L, Barnett JS, Tucker M,
Hardarson T. Cross-validation and predictive value of near-
infrared spectroscopy algorithms for day-5 blastocyst transfer.
Reprod Biomed Online 2011; 22:477–484.
171. Seli E, Bruce C, Botros L, Henson M, Roos P, Judge K, Hardarson
T, Ahlstrom A, Harrison P, Henman M, Go K, Acevedo N et al.
Receiver operating characteristic (ROC) analysis of day 5 mor-
phology grading and metabolomic viability score on predicting
implantation outcome. J Assist Reprod Genet 2011; 28:137–144.
172. Vergouw CG, Botros LL, Judge K, Henson M, Roos P, Kostelijk
EH, Schats R, Twisk JW, Hompes PG, Sakkas D, Lambalk CB.
Non-invasive viability assessment of day-4 frozen-thawed human
embryos using near infrared spectroscopy. Reprod Biomed
Online 2011; 23:769–776.
173. Kirkegaard K, Svane AS, Nielsen JS, Hindkjaer JJ, Nielsen NC,
Ingerslev HJ. Nuclear magnetic resonance metabolomic profiling
of day 3 and 5 embryo culture medium does not predict preg-
nancy outcome in good prognosis patients: a prospective cohort
study on single transferred embryos. Hum Reprod 2014; 29:
2413–2420.
174. Hardarson T, Ahlstrom A, Rogberg L, Botros L, Hillensjo T,
Westlander G, Sakkas D, Wikland M. Non-invasive metabolomic
profiling of day 2 and 5 embryo culture medium: a prospective
randomized trial. Hum Reprod 2012; 27:89–96.
175. Vergouw CG, Kieslinger DC, Kostelijk EH, Botros LL, Schats R,
Hompes PG, Sakkas D, Lambalk CB. Day 3 embryo selection by
metabolomic profiling of culture medium with near-infrared spec-
troscopy as an adjunct to morphology: a randomized controlled
trial. Hum Reprod 2012; 27:2304–2311.
176. Sfontouris, Lainas GT, Sakkas D, Zorzovilis IZ, Petsas GK, Lainas
TG. Non-invasive metabolomic analysis using a commercial NIR
instrument for embryo selection. J Hum Reprod Sci 2013; 6:
133–139.
177. Bracewell-Milnes T, Saso S, Abdalla H, Nikolau D, Norman-
Taylor J, Johnson M, Holmes E, Thum MY. Metabolomics as a
tool to identify biomarkers to predict and improve outcomes in
reproductive medicine: a systematic review. Hum Reprod Update
2017; 23:723–736.
178. Song J, Wang X, Guo Y, Yang Y, Xu K, Wang T, Sa Y,
Yuan L, Jiang H, Guo J, Sun Z. Novel high-coverage targeted
metabolomics method (SWATHtoMRM) for exploring follicular
fluid metabolome alterations in women with recurrent sponta-
neous abortion undergoing in vitro fertilization. Sci Rep 2019;
9:10873.
179. Urbanski JP, Johnson MT, Craig DD, Potter DL, Gardner DK,
Thorsen T. Noninvasive metabolic profiling using microfluidics
for analysis of single preimplantation embryos. Anal Chem 2008;
80:6500–6507.
180. Heo YS, Cabrera LM, Bormann CL, Smith GD, Takayama S.
Real time culture and analysis of embryo metabolism using a
microfluidic device with deformation based actuation. Lab Chip
2012; 12:2240–2246.
181. Meseguer M, Herrero J, Tejera A, Hilligsoe KM, Ramsing NB,
Remohi J. The use of morphokinetics as a predictor of embryo
implantation. Hum Reprod 2011; 26:2658–2671.
182. Wong CC, Loewke KE, Bossert NL, Behr B, De Jonge CJ, Baer
TM, Reijo Pera RA. Non-invasive imaging of human embryos
before embryonic genome activation predicts development to the
blastocyst stage. Nat Biotechnol 2010; 28:1115–1121.
183. Chavez SL, Loewke KE, Han J, Moussavi F, Colls P, Munne S,
Behr B, Reijo Pera RA. Dynamic blastomere behaviour ref lects
human embryo ploidy by the four-cell stage. Nat Commun 2012;
3:1251.
184. Racowsky C, Kovacs P, Martins WP. A critical appraisal of
time-lapse imaging for embryo selection: where are we and
where do we need to go? J Assist Reprod Genet 2015; 32:
1025–1030.
185. Gardner DK, Balaban B. Assessment of human embryo devel-
opment using morphological criteria in an era of time-lapse,
algorithms and ’OMICS’: is looking good still important? Mol
Hum Reprod 2016; 22:704–718.
186. Ottosen LD, Hindkjaer J, Ingerslev J. Light exposure of the ovum
and preimplantation embryo during ART procedures. J Assist
Reprod Genet 2007; 24:99–103.
187. Jasensky J, Swain JE. Peering beneath the surface: novel imaging
techniques to noninvasively select gametes and embryos for ART.
Biol Reprod 2013; 89:105.
188. Swann MM, Mitchison JM. Refinements in polarized light
microscopy. J Exp Biol 1950; 27:226–237.
189. Keefe D, Tran P, Pellegrini C, Oldenbourg R. Polarized light
microscopy and digital image processing identify a multilaminar
structure of the hamster zona pellucida. Hum Reprod 1997; 12:
1250–1252.
190. Silva CP, Kommineni K, Oldenbourg R, Keefe DL. The first polar
body does not predict accurately the location of the metaphase
II meiotic spindle in mammalian oocytes. Fertil Steril 1999; 71:
719–721.
191. Gomes CM, Silva CA, Acevedo N, Baracat E, Serafini P, Smith
GD. Influence of vitrification on mouse metaphase II oocyte
spindle dynamics and chromatin alignment. Fertil Steril 2008; 90:
1396–1404.
192. Squirrell JM, Wokosin DL, White JG, Bavister BD. Long-
term two-photon fluorescence imaging of mammalian embryos
without compromising viability. Nat Biotechnol 1999; 17:
763–767.
193. Wan Y, McDole K, Keller PJ. Light-sheet microscopy and its
potential for understanding developmental processes. Annu Rev
Cell Dev Biol 2019; 35:655–681.
194. Albert-Smet I, Marcos-Vidal A, Vaquero JJ, Desco M, Munoz-
Barrutia A, Ripoll J. Applications of light-sheet microscopy in
microdevices. Front Neuroanat 2019; 13:1.
195. Wood BR, Chernenko T, Matthaus C, Diem M, Chong C,
Bernhard U, Jene C, Brandli AA, McNaughton D, Tobin MJ,
Trounson A, Lacham-Kaplan O. Shedding new light on the molec-
ular architecture of oocytes using a combination of synchrotron
Fourier transform-infrared and Raman spectroscopic mapping.
Anal Chem 2008; 80:9065–9072.
196. Rusciano G, Pesce G, Salemme M, Selvaggi L, Vaccaro C, Sasso
A, Carotenuto R. Raman spectroscopy of Xenopus laevis oocytes.
Methods 2010; 51:27–36.
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
R. Sciorio et al. 17
197. Bogliolo L, Murrone O, Di Emidio G, Piccinini M, Ariu F, Ledda
S, Tatone C. Raman spectroscopy-based approach to detect aging-
related oxidative damage in the mouse oocyte. J Assist Reprod
Genet 2013; 30:877–882.
198. Bogliolo L, Ledda S, Innocenzi P, Ariu F, Bebbere D, Rosati
I, Leoni GG, Piccinini M. Raman microspectroscopy as a non-
invasive tool to assess the vitrification-induced changes of ovine
oocyte zona pellucida. Cryobiology 2012; 64:267–272.
199. Jasensky J, Boughton AP, Khmaladze A, Ding J, Zhang C, Swain
JE, Smith GW, Chen Z, Smith GD. Live-cell quantification and
comparison of mammalian oocyte cytosolic lipid content between
species, during development, and in relation to body composi-
tion using nonlinear vibrational microscopy. Analyst 2016; 141:
4694–4706.
200. Angione SL, Oulhen N, Brayboy LM, Tripathi A, Wessel GM.
Simple perfusion apparatus for manipulation, tracking, and study
of oocytes and embryos. Fertil Steril 2015; 103:281–290 e285.
201. Reich A, Klatsky P, Carson S, Wessel G. The transcriptome of
a human polar body accurately reflects its sibling oocyte. JBiol
Chem 2011; 286:40743–40749.
202. Hou Y, Fan W, Yan L, Li R, Lian Y, Huang J, Li J, Xu L, Tang F,
Xie XS, Qiao J. Genome analyses of single human oocytes. Cell
2013; 155:1492–1506.
203. Wolf DP, Mitalipov N, Mitalipov S. Mitochondrial replacement
therapy in reproductive medicine. Trends Mol Med 2015; 21:
68–76.
204. Huang L, Bogale B, Tang Y, Lu S, Xie XS, Racowsky
C. Noninvasive preimplantation genetic testing for aneu-
ploidy in spent medium may be more reliable than tro-
phectoderm biopsy. Proc Natl Acad Sci U S A 2019; 116:
14105–14112.
205. Leaver M, Wells D. Non-invasive preimplantation genetic test-
ing (niPGT): the next revolution in reproductive genetics? Hum
Reprod Update 2020; 26:16–42.
Downloaded from https://academic.oup.com/biolreprod/advance-article/doi/10.1093/biolre/ioac009/6523151 by guest on 07 February 2022
... Despite improvements in culture media, the introduction of intracytoplasmic sperm injection (ICSI) [14] and embryo and oocyte vitrification [15], limited progress has been made in oocyte quality assessments. There is no intra or interlaboratory standardization of oocyte grading amongst embryologists therefore leading to high subjectivity [16,17]. ...
... Embryologists utilize light microscopy to notate extracytoplasmic and cytoplasmic abnormalities when visualizing oocytes for intracytoplasmic sperm injection [17,25,31,32]. Oocyte abnormalities include unusual zona pellucida (ZP) or the extracellular matrix surrounding an oocyte. ...
... Confocal Raman spectroscopy Confocal Raman spectroscopy (CRS) employs inelastic scattering to produce interactions of light and matter. Photon scattering allows for identification of molecules and the molecular bonds of cells [17]. Although CRS is non-invasive, it does require long imaging times of live cells which could be detrimental to oocytes retrieved during IVF. ...
Article
Full-text available
Determining egg quality is the foremost challenge in assisted reproductive technology (ART). Although extensive advances have been made in multiple areas of ART over the last forty years, oocyte quality assessment tools have not much evolved beyond standard morphological observation. The oocyte not only delivers half of the nuclear genetic material and all of the mitochondrial DNA to an embryo but also provides complete developmental support during embryonic growth. Oocyte mitochondrial numbers far exceed those of any somatic cell, yet little work has been done to evaluate the mitochondrial bioenergetics of an oocyte. Current standard oocyte assessment in in vitro fertilization (IVF) centers include the observation of oocytes and their surrounding cell complex (cumulus cells) via stereomicroscope or inverted microscope, which is largely primitive. Additional oocyte assessments include polar body grading and polarized light meiotic spindle imaging. However, the evidence regarding the aforementioned methods of oocyte quality assessment and IVF outcomes is contradictory and non-reproducible. High resolution microscopy techniques have also been implemented in animal and human models with promising outcomes. The current era of oocyte imaging continues to evolve with discoveries in artificial intelligence models of oocyte morphology selection albeit at a slow rate. In this review, past, current, and future oocyte imaging techniques will be examined with the goal of drawing attention to the gap which limits our ability to assess oocytes in real time. The implications of improved oocyte imaging techniques on patients undergoing IVF will be discussed as well as the need to develop point of care oocyte assessment testing in IVF labs.
... In red blood cells, advancements in exploring RBC deformability changes induced by P. falciparum and understanding the biophysical parameters regulating their bio-rheological behavior have significantly improved in recent decades [10]. Given the critical changes in gamete and early embryo viscoelasticity, it is logical that alterations in the biomechanical properties of oocytes or embryos may offer insight into their fertilization, embryo viability, genetic abnormality, pregnancy [11][12][13][14]. To date, there is no standard guideline or technique to aid in the selection of oocytes that have higher likelihood of developing into usable blastocysts, which is chosen for transfer or vitrification. ...
... Several studies indicated that alterations in oocytes stiffness may affect fertilization, embryo viability, genetic abnormality, and pregnancy [11][12][13][14], suggesting that viscoelasticity could serve as an indicator in the aspiration measurement prior to ICSI or classical IVF procedure. However, a more stringent and well-designed study will be required to establish this as for proof-of-concept. ...
Article
Full-text available
Purpose The purposes of this study were to determine whether biomechanical properties of mature oocytes could predict usable blastocyst formation better than morphological information or maternal factors, and to demonstrate the safety of the aspiration measurement procedure used to determine the biomechanical properties of oocytes. Methods A prospective split cohort study was conducted with patients from two IVF clinics who underwent in vitro fertilization. Each patient’s oocytes were randomly divided into a measurement group and a control group. The aspiration depth into a micropipette was measured, and the biomechanical properties were derived. Oocyte fertilization, day 3 morphology, and blastocyst development were observed and compared between measured and unmeasured cohorts. A predictive classifier was trained to predict usable blastocyst formation and compared to the predictions of four experienced embryologists. Results 68 patients and their corresponding 1252 oocytes were included in the study. In the safety analyses, there was no significant difference between the cohorts for fertilization, while the day 3 and 5 embryo development were not negatively affected. Four embryologists predicted usable blastocyst development based on oocyte morphology with an average accuracy of 44% while the predictive classifier achieved an accuracy of 71%. Retaining the variables necessary for normal fertilization, only data from successfully fertilized oocytes were used, resulting in a classifier an accuracy of 81%. Conclusions To date, there is no standard guideline or technique to aid in the selection of oocytes that have a higher likelihood of developing into usable blastocysts, which are chosen for transfer or vitrification. This study provides a comprehensive workflow of extracting biomechanical properties and building a predictive classifier using these properties to predict mature oocytes’ developmental potential. The classifier has greater accuracy in predicting the formation of usable blastocysts than the predictions provided by morphological information or maternal factors. The measurement procedure did not negatively affect embryo culture outcomes. While further analysis is necessary, this study shows the potential of using biomechanical properties of oocytes to predict embryo developmental outcomes.
... In addition, the level of experience and physical abilities such as vision, and mental and physical agility of the investigator can affect the decision about the quality of oocytes. Furthermore, a significant percentage of embryos fail implantation, even though they are morphologically rated as high-quality embryos by embryologists or even after genetically analysis [18]. As a result, routine morphological ocular classification cannot be considered a fail-proof and comprehensive method of oocyte quality determination. ...
... In recent years, articles on routine morphological methods of oocyte quality evaluation and even genetic screening and other biological-based evaluations have been published that provide in-depth assessment of the morphology and biology of the first polar body, zona, cumulus cells, follicles, follicular fluid, and environment [18]. However, since these developments are not strictly under the category of engineering, they are not discussed in this paper. ...
Article
Full-text available
Although assisted reproductive technology has been very successful for the treatment of infertility, its steps are still dependent on direct human opinion. An important step of assisted reproductive treatments in lab for women is choosing an oocyte that has a better quality. This step would predict which oocyte has developmental competence leading to healthy baby. Observation of the oocyte morphological quality indicators under microscope by an embryologist is the most common evaluation method of oocyte quality. Such subjective method which relies on embryologist’s experience may vary and leads to misdiagnosis. An alternative solution to eliminate human misjudging in traditional methods and overcome the limitations of them is always using engineering-based procedure. In this review article, was deep studied and categorized engineering-based methods applied for the evaluation of oocyte quality. Then, the challenges in laboratories and clinics settings move forward with translational medicine perspective in mind for all those methods which had been studied were discussed. Finally, a standardized process was presented may help improving and focusing research in this field. Moreover, effective suggestion techniques were introduced that are expect they would be complementary methods to accelerate future researches. The aim of this review was to create a new prospect with the engineering approaches to evaluate oocyte quality and we hope this would help infertile couples to get a baby.
... On day 4, the embryo compacts, resulting in the formation of the 'morula' a groups of blastomeres. On day 5, the embryo become a 'blastocyst', a fluid-filled structure formed of the inner cell mass (white arrowhead) and trophectoderm (grey arrowhead) the late 1990s with the consistency achieved from these commercially produced media, it became more common to culture embryos to the blastocyst stage, resulting in an improvement in pregnancy rate and reduction of multiple pregnancies with the transfer of single blastocyst [1,2,[12][13][14][15][16][17]. ...
Article
Full-text available
In the last four decades, the assisted reproductive technology (ART) field has witnessed advances, resulting in improving pregnancy rates and diminishing complications, in particular reduced incidence of multiple births. These improvements are secondary to advanced knowledge on embryonic physiology and metabolism, resulting in the ability to design new and improved culture conditions. Indeed, the incubator represents only a surrogate of the oviduct and uterus, and the culture conditions are only imitating the physiological environment of the female reproductive tract. In vivo, the embryo travels through a dynamic and changing environment from the oviduct to the uterus, while in vitro, the embryo is cultured in a static fashion. Importantly, while culture media play a critical role in optimising embryo development, a large host of additional factors are equally important. Additional potential variables, including but not limited to pH, temperature, osmolality, gas concentrations and light exposure need to be carefully controlled to prevent stress and permit optimal implantation potential. This manuscript will provide an overview of how different current culture conditions may affect oocyte and embryo viability with particular focus on human literature.
... Analysis of biofluids has been investigated as a non-invasive marker to assess oocyte competence in human-assisted reproduction [22], so the measurement of metabolic byproducts in spent culture media has the potential to be a non-invasive marker of the health of follicles grown in vitro. The application of metabolomics to ovine-isolated secondary follicles using a spent culture medium has been attempted, but no clear conclusions could be drawn [23]. ...
Article
Full-text available
The amino acid metabolism of bovine follicles during in vitro growth (IVG) was evaluated to identify potential indicators of health during culture. The bovine ovarian cortex was sliced, prepared as strips, and cultured for 6 days. Tissue samples were examined histologically before and after 6 days of culture, and the degree of follicle activation was classified as either high or low based on the number of growing secondary follicles present (high: 7~11; low: 0~1). In a separate experiment, secondary follicles (diameter range: 100~200 μm) were manually isolated and cultured, and their growth was monitored for 6 days. Cultured follicles were classified as growth or degenerate based on diameter change during culture (growth: +60.5~74.1 μm; degenerate: −28~15.2 μm). Free amino acids and their metabolites were measured in the spent culture medium from each group. In cultured ovarian cortical strips, the concentration of α-aminoadipic acid was significantly higher in the low activation group than in the high group (p < 0.05), while those of methionine, lysine, and arginine were higher in the high activation group. In cultured isolated secondary follicles, concentrations of methionine, tyrosine, histidine, and hydroxyproline were higher in the degenerate group (p ≤ 0.05). In conclusion, amino acid metabolism has the potential to serve as an indicator of primordial follicle activation and subsequent growth rate during bovine IVG.
... Because of the difficulty in evaluating oocyte quality in patients undergoing IVF [19][20][21], the cumulative live birth rate was used to observe the clinical efficacy, which is the most critical and valuable indicator to evidence the clinical outcome [22,23]. In this study, we aimed to assess whether GH addition as an adjuvant therapy could increase the cumulative live birth rate in patients with poor embryo quality in the previous IVF cycles. ...
Article
Full-text available
This study aimed to assess the effects of GH adjuvant therapy on the cumulative live birth rate in patients with poor embryo quality and to determine the characteristics of patients who are more responsive to GH. A retrospective cohort study was carried out in patients who have suffered from previous IVF failure due to poor embryonic development and underwent IVF with or without a 6-week pretreatment with GH in the subsequent cycle from January 2018 to December 2020. Clinical parameters including the cumulative live birth rate between the (−) GH and (+) GH groups were compared. Multivariate analysis was performed to ascertain associations between clinical parameters and cumulative live birth rate. Upon analysis of the clinical data from 236 IVF cycles, 84 patients received GH and 152 did not receive GH. In frozen embryo transfer cycles, compared with the (−) GH group, the implantation rate and live birth rate were significantly higher in the (+) GH group (p < 0.05). After adjusting for possible confounding factors, GH improved cumulative live birth per oocyte retrieval cycle by 1.96 folds (p = 0.032). Furthermore, when patients were subdivided based on age and BMI, a significant increase in the cumulative live birth rate was found in the (+) GH group of patients between 35 and 42 years old and BMI ≥ 24 kg/m2, respectively (p < 0.05). GH may increase the live birth rate in women who experienced IVF failure because of poor embryonic development, particularly in obese patients and women with advanced age.
Article
Despite significant advancements in assisted reproductive technology, there is still high demand for improvements in the clinical pregnancy rate and reductions in the time needed to achieve a live birth. One bottleneck issue is the effective selection of oocytes and/or embryos of better quality in the laboratory. The quality of oocytes and embryos cannot be accurately assessed with routine clinical methods. Thus, assessment methods that are more objective, accurate, quantifiable, non-invasive, rapid, and repeatable are needed in the clinic. This perspective highlights the latest developments in non-invasive assessments of oocyte and embryo quality and presents new trends and insights into the biomedical engineering technologies used to evaluate oocyte and embryo quality.
Preprint
Background Egg quality is a limiting factor of female fertility and assisted reproductive technology (ART) success. Oocytes recovered from hyperstimulated ovaries often display morphological anomalies suspected to compromise their fertilization and developmental potential. Knowledge of (ab)normal oocyte’s intracellular organization is vital to establish reliable criteria for morphological evaluation of oocytes intended for in vitro fertilization (IVF). Methods Transmission electron microscopy (TEM) was used to investigate the fine morphology of 22 dysmorphic IVF eggs exhibiting different types of cytoplasmic irregularities, namely (1) refractile bodies, (2) centrally-located cytoplasmic granularity (CLCG), (3) smooth endoplasmic reticulum (SER) disc, and (4) vacuoles. The cytoskeleton targeting compounds were employed to address the causative mechanism behind the anomalous cytoplasmic architecture observed in abnormal egg samples. A total of 133 immature oocytes were exposed to chemical inhibitors/control conditions, and their morphology was examined by fluorescent and electron microscopy. Results TEM exposed the structural basis of the common oocyte aberrations and revealed that the underlying cause of two of the studied morphotypes was excessive organelle clustering. Inhibition experiments showed that disruption of actin, not microtubules, allows inordinate aggregation of subcellular structures resembling the ultrastructural pattern seen in morphologically abnormal eggs retrieved in IVF cycles. These results imply that actin serves as a regulator of organelle distribution during human oocyte maturation. Conclusions The ultrastructural analogy between dysmorphic eggs and oocytes, in which actin network integrity was perturbed, suggests that malfunction of the actin cytoskeleton might be implicated in generating common cytoplasmic aberrations. Knowledge of human oocytes’ inner workings and the origin of morphological abnormalities is a step forward to more objective egg quality assessment in clinical practice.
Article
Full-text available
Meiosis is the basis of sexual reproduction. In female mammals, meiosis of oocytes starts before birth and sustains at the dictyate stage of meiotic prophase I before gonadotropins-induced ovulation happens. Once meiosis gets started, the oocytes undergo the leptotene, zygotene, and pachytene stages, and then arrest at the dictyate stage. During each estrus cycle in mammals, or menstrual cycle in humans, a small portion of oocytes within preovulatory follicles may resume meiosis. It is crucial for females to supply high quality mature oocytes for sustaining fertility, which is generally achieved by fine-tuning oocyte meiotic arrest and resumption progression. Anything that disturbs the process may result in failure of oogenesis and seriously affect both the fertility and the health of females. Therefore, uncovering the regulatory network of oocyte meiosis progression illuminates not only how the foundations of mammalian reproduction are laid, but how mis-regulation of these steps result in infertility. In order to provide an overview of the recently uncovered cellular and molecular mechanism during oocyte maturation, especially epigenetic modification, the progress of the regulatory network of oocyte meiosis progression including meiosis arrest and meiosis resumption induced by gonadotropins is summarized. Then, advances in the epigenetic aspects, such as histone acetylation, phosphorylation, methylation, glycosylation, ubiquitination, and SUMOylation related to the quality of oocyte maturation are reviewed.
Article
Full-text available
Female fertility relies on successful egg development. Besides chromosome segregation, complex structural and biochemical changes in the cytoplasmic compartment are necessary to confer the female gamete the capacity to undergo normal fertilization and sustain embryonic development. Despite the profound impact on egg quality, morphological bases of cytoplasmic maturation remain largely unknown. Here, we report our findings from the ultrastructural analysis of 69 unfertilized human oocytes from 34 young and healthy egg donors. By comparison of samples fixed at three consecutive developmental stages, we explored how ooplasmic architecture changes during meiotic maturation in vitro. The morphometric image analysis supported observation that the major reorganization of cytoplasm occurs before polar body extrusion. The organelles initially concentrated around prophase nucleus were repositioned toward the periphery and evenly distributed throughout the ooplasm. As maturation progressed, distinct secretory apparatus appeared to transform into cortical granules that clustered underneath the oocyte's surface. The most prominent feature was the gradual formation of heterologous complexes composed of variable elements of endoplasmic reticulum and multiple mitochondria with primitive morphology. Based on the generated image dataset, we proposed a morphological map of cytoplasmic maturation, which may serve as a reference for future comparative studies. In conclusion, this work improves our understanding of human oocyte morphology, cytoplasmic maturation, and intracellular factors defining human egg quality. Although this analysis involved spare oocytes completing development in vitro, it provides essential insight into the enigmatic process by which human egg progenitors prepare for fertilization.
Article
Full-text available
The complexity of follicular fluid metabolome presents a significant challenge for qualitative and quantitative metabolite profiling, and for discovering the comprehensive biomarkers. In order to address this challenge, a novel SWATHtoMRM metabolomics method was used for providing broad coverage and excellent quantitative capability to discover the human follicular fluid metabolites related to recurrent spontaneous abortion (RSA) after in vitro fertilization and embryo transfer, and to evaluate their relationship with pregnancy outcome. The follicular fluid samples from the spontaneous abortion group (n = 22) and the control group (n = 22) were analyzed using ultra-performance liquid chromatography high-resolution mass spectrometry. A novel, high-coverage, targeted metabolomics method (SWATH to MRM) and a targeted metabolomics method were used to find and validate the differential metabolites between the two groups. A total of 18 follicular fluid metabolites, including amino acids, cholesterol, vitamins, fatty acids, cholic acid, lysophosphatidylcholine and other metabolites, were identified. In the RSA group, 8 metabolites, namely dehydroepiandrosterone, lysoPC(16:0), lysoPC(18:2), lysoPC(18:1), lysoPC(18:0), lysoPC(20:5), lysoPC(20:4), and lysoPC(20:3), were up-regulated, and 10 metabolites, namely phenylalanine, linoleate, oleic acid, docosahexaenoic acid, lithocholic acid, 25-hydroxyvitamin D3, hydroxycholesterol, 13-hydroxy-alpha-tocopherol, leucine, and tryptophan, were down-regulated. These differential metabolites related to RSA may provide a possible diagnostic basis and therapeutic target for RSA, as well as a scientific basis for elucidating the mechanism of RSA.
Article
Full-text available
Significance In clinical in vitro fertilization (IVF), the prevailing method for preimplantation genetic testing for aneuploidy (PGT-A) requires biopsying a few cells from the trophectoderm (TE), the lineage that forms the placenta. The test is invasive, requires specialized skills, and suffers from false positives and negatives because chromosome numbers in the TE and the inner cell mass (ICM), which develops into the fetus, are not always the same. Noninvasive PGT-A (niPGT-A), which is based on sequencing DNA released into the culture medium from both TE and ICM, may offer a solution to these problems but has previously had limited efficacy. The present results show improved sensitivity and reliability of niPGT-A, suggesting a potentially superior test for noninvasive and cost-effective PGT-A in clinical IVF.
Article
Full-text available
Background: Human follicular fluid is an intricate biological fluid contributing to the developing oocyte microenvironment. Accumulating evidence suggests that sex hormones present in follicular fluid (FF) may play an important role in regulating oocyte developmental potential. The aim of this study was to determine if anti-Müllerian hormone (AMH) and progesterone (P4) levels in FF are correlated with oocyte quality as defined by subsequent embryonic development. Methods: This was a prospective cohort study of 88 women undergoing IVF/ICSI at a university associated fertility clinic. Follicular fluid was collected from the first follicle aspirated at the time of oocyte retrieval. The corresponding oocyte was individually cultured in order to track its developmental outcome. FF-AMH and P4 concentrations from follicles where the oocyte fertilised normally and developed into a blastocyst on day 5 (Group 1: BLAST, n = 23) were compared with FF from follicles where the oocyte fertilised normally but failed to reach blastocyst stage by day 5 (Group 2: FERT, n = 19). No significant differences were observed between the two groups in terms of maternal age, body mass index, previous live births, previous pregnancy loss, number of antral follicles, number of oocytes recovered, IVF:ICSI ratio or percentage of recovered oocytes that fertilised. Results: FF-AMH and P4 levels were significantly increased in Group 1: BLAST compared to Group 2: FERT (P = 0.007 and P = 0.013 respectively). Twenty-one FF samples had an AMH level > 15 pmol/L, of which 17 related to oocytes that progressed to blastocyst stage, providing a positive prediction value (PPV) of 76.96%. Eleven FF samples had a P4 level > 60 mg/ml, of which 10 progressed to blastocyst stage, providing a PPV of 90.99%. Six samples had an AMH level > 15 pmol/L and a P4 level > 60 mg/ml, of which 100% progressed to blastocyst stage, providing a PPV of 96.83%. Conclusions: FF-AMH and P4 levels from individual follicles can accurately predetermine subsequent embryonic development. Combining follicular fluid analysis with routine morphological assessment, could allow for a more accurate and sensitive method of determining embryonic developmental competence.
Article
Full-text available
Drug of abuse (DOA) consumption is a growing problem worldwide, particularly with increasing numbers of new psychoactive substances (NPS) entering the drug market. Generally, little information on their adverse effects and toxicity are available. The direct detection and identification of NPS is an analytical challenge due to their ephemerality on the drug scene. An approach that does not directly focus on the structural detection of an analyte or its metabolites, would be beneficial for this complex analytical scenario and the development of alternative screening methods could help to provide fast response on suspected NPS consumption. A metabolomics approach might represent such an alternative strategy for the identification of biomarkers for different questions in DOA testing. Metabolomics is the monitoring of changes in small (endogenous) molecules (<1,000 Da) in response to a certain stimulus, e.g., DOA consumption. For this review, a literature search targeting “metabolomics” and different DOAs or NPS was conducted. Thereby, different applications of metabolomic strategies in biomarker research for DOA identification were identified: (a) as an additional tool for metabolism studies bearing the major advantage that particularly a priori unknown or unexpected metabolites can be identified; and (b) for identification of endogenous biomarker or metabolite patterns, e.g., for synthetic cannabinoids or also to indirectly detect urine manipulation attempts by chemical adulteration or replacement with artificial urine samples. The majority of the currently available literature in that field, however, deals with metabolomic studies for DOAs to better assess their acute or chronic effects or to find biomarkers for drug addiction and tolerance. Certain changes in endogenous compounds are detected for all studied DOAs, but often similar compounds/pathways are influenced. When evaluating these studies with regard to possible biomarkers for drug consumption, the observed changes appear, albeit statistically significant, too small to reliably work as biomarker for drug consumption. Further, different drugs were shown to affect the same pathways. In conclusion, metabolomic approaches possess potential for detection of biomarkers indicating drug consumption. More studies, including more sensitive targeted analyses, multi-variant statistical models or deep-learning approaches are needed to fully explore the potential of omics science in DOA testing.
Article
Background: Preimplantation genetic testing (PGT) encompasses methods that allow embryos to be tested for severe inherited conditions or for chromosome abnormalities, relevant to embryo health and viability. In order to obtain embryonic genetic material for analysis, a biopsy is required, involving the removal of one or more cells. This invasive procedure greatly increases the costs of PGT and there have been concerns that embryo viability could be compromised in some cases. The recent discovery of DNA within the blastocoele fluid (BF) of blastocysts and in spent embryo culture media (SCM) has led to interest in the development of non-invasive methods of PGT (niPGT). Objective and rationale: This review evaluates the current scientific evidence regarding non-invasive genetic assessment of preimplantation embryos. The success of different PGT methodologies in collecting and analysing extra-embryonic DNA is evaluated, and consideration is given to the potential biological and technical hindrances to obtaining a reliable clinical diagnosis. Search methods: Original research and review papers concerning niPGT were sourced by searching PubMed and Google Scholar databases until July 2019. Searches comprised the keywords: 'non-invasive'; 'cell-free DNA'; 'blastocentesis'; 'blastocoel fluid'; 'spent culture media'; 'embryo culture medium'; 'preimplantation genetic testing'; 'preimplantation genetic diagnosis'; 'preimplantation genetic screening'; and 'aneuploidy'. Outcomes: Embryonic DNA is frequently detectable in BF and SCM of embryos produced during IVF treatment. Initial studies have achieved some success when performing cytogenetic and molecular genetic analysis. However, in many cases, the efficiency has been restricted by technical complications associated with the low quantity and quality of the DNA. Reported levels of ploidy agreement between SCM/BF samples and biopsied embryonic cells vary widely. In some cases, a discrepancy with respect to cytogenetic data obtained after trophectoderm biopsy may be attributable to embryonic mosaicism or DNA contamination (usually of maternal origin). Some research indicates that aneuploid cells are preferentially eliminated from the embryo, suggesting that their DNA might be over-represented in SCM and BF samples; this hypothesis requires further investigation. Wider implications: Available data suggest that BF and SCM samples frequently provide DNA templates suitable for genetic analyses, offering a potential means of PGT that is less expensive than traditional methods, requires less micromanipulation skill and poses a lower risk to embryos. Critically, DNA isolation and amplification protocols must be optimised to reproducibly obtain an accurate clinical diagnosis, whilst minimising the impact of confounding factors such as contamination. Further investigations are required to understand the mechanisms underlying the release of embryonic DNA and to determine the extent to which this material reflects the true genetic status of the corresponding embryo. Currently, the clinic al potential of niPGT remains unknown.
Article
Objective: To determine if natural selection and follicular stimulation produces a lower risk for embryonic aneuploidy than that attained following superovulation with exogenous gonadotropins. Design: Prospective observational with historical control group. Setting: Large academically affiliated private practice. Patient(s): All patients presenting for their evaluation for infertility were offered participation in the study. Intervention(s): All participants in the natural cycle group underwent an unstimulated in vitro fertilization (IVF) cycle. A subsequent frozen embryo transfer was performed if a euploid blastocyst was attained. Main outcome measure(s): Rates of embryonic aneuploidy attained in unstimulated IVF cycles were compared to those observed in age-controlled historical cohort undergoing conventional stimulated IVF cycles with exogenous gonadotropins. Result(s): Aneuploidy rates were equivalent in unstimulated and stimulated IVF cycles. The prevalence of aneuploidy in natural cycles increased with the age of the female partner in a manner identical to that seen in stimulated IVF cycles. Finally, sustained implantation rates of euploid blastocysts were equivalent in natural and stimulated IVF cycles. Conclusion(s): Rates of embryonic aneuploidy are not impacted by follicular stimulation with exogenous gonadotropins. Prior concerns of inducing a higher risk of embryonic aneuploidy are not supported by this data. Clinical trial registration number: NCT01866618.
Article
The ability to visualize and quantitatively measure dynamic biological processes in vivo and at high spatiotemporal resolution is of fundamental importance to experimental investigations in developmental biology. Light-sheet microscopy is particularly well suited to providing such data, since it offers exceptionally high imaging speed and good spatial resolution while minimizing light-induced damage to the specimen. We review core principles and recent advances in light-sheet microscopy, with a focus on concepts and implementations relevant for applications in developmental biology. We discuss how light-sheet microcopy has helped advance our understanding of developmental processes from single-molecule to whole-organism studies, assess the potential for synergies with other state-of-the-art technologies, and introduce methods for computational image and data analysis. Finally, we explore the future trajectory of light-sheet microscopy, discuss key efforts to disseminate new light-sheet technology, and identify exciting opportunities for further advances. Expected final online publication date for the Annual Review of Cell and Developmental Biology Volume 35 is October 7, 2019. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Multiple pregnancies with higher risk of preterm birth and the associated higher morbidity have been a major obstacle from the early days of in vitro fertilization. A good strategy to avoid multiple pregnancies is elective single embryo transfer and cryopreservation of spare embryos. Important factors in adopting this strategy are good counselling of the patients and the selection of embryos with high implantation potential. Technical advances in embryo selection have been described during recent years, time lapse monitoring and genetic assessment of the embryos being the most important achievements. With these studies we have gained new information on early embryos. However, at present, there is insufficient evidence to recommend the routine use of these new techniques. The ultimate goal of infertility treatment is a healthy baby.