Вы находитесь на странице: 1из 13

CSIRO PUBLISHING

www.publish.csiro.au/journals/rfd Reproduction, Fertility and Development, 2007, 19, 719–731

Novel gamete storage

A. DinnyesA,C , J. LiuA and T. L. NedambaleB


A BiotalentumLtd, Aulich L. 26, Godollo 2100, Hungary.
BAnimal Production Institute, Indigenous Genotype Physiology and Biotechnology
Development, Private Bag X2, Irene 0062, South Africa.
C Corresponding author. Email: biotalentum@yahoo.com

Abstract. The aim of this review is to outline recent advances in gamete storage that are beneficial for rescuing endangered
species or for the breeding of companion animals. Much more information is available on the technical resolutions and
practical applications of sperm cryopreservation in various species than of female gametes, reproductive tissues or organs.
Mammalian sperm cryopreservation often works relatively efficiently; however, the ability of female gametes to be
cryopreserved and still be viable for fertilisation is also essential for rescuing endangered species. For a proper evaluation
of gamete cryopreservation possibilities in a given species, it is essential to understand the basic mechanism affecting the
survival of cryopreserved cells, the technical and physical limitations, the available techniques and the new avenues to
resolve the specific problems in that species. This paper is aimed to provide some help for this process. The limited length
of this paper resulted in the omission of information on many important areas, including most data on teleosts, amphibian
and insect cryopreservation.

Current status of knowledge Polge et al. (1949) successfully cryopreserved bull sperma-
The roots of modern applied gamete storage date back more tozoa by introducing glycerol as a cryoprotective agent (CPA)
than 50 years to the time when an efficient cryopreservation into freezing medium for the first time. Later many other
technique for preserving spermatozoa with glycerol was acci- non-electrolytic permeating CPA, such as dimethyl sulfoxide,
dentally discovered (Polge et al. 1949). As a result, serious ethylene glycol and propylene glycol, were introduced for other
development began and sperm cryopreservation has become cell types (such as embryos and oocytes) in various species
an important method in agriculture and medicine and also (Mazur 1970).
an option for rescuing endangered species. To some extent Although there have been some recent exciting studies involv-
gamete storage can provide insurance for preserving the exist- ing freeze drying and vitrification (see later section) of sper-
ing genetic diversity within endangered species. These species matozoa as a means of conserving this genetic material, slow
survive in nature but often in fragmented habitats, or live in equilibrium freezing is still the most broadly practiced sperm
geographically disparate zoos and they are also susceptible to cryopreservation method. It consists of the following series of
inbreeding depression, environmental catastrophes, epidemics, non-physiological steps:
and even to drastic shifts in social and political structures (Wildt
(1) adding a permeating CPA (e.g. glycerol, ethylene glycol,
2000; Wildt et al. 2001; Pukazhenthi et al. 2006a). However,
dimethyl sulfoxide, propylene glycol) or non-permeating
there have been many encouraging results following artificial
CPA (e.g. sucrose, glucose, fructose, sorbitol, trehalose or
insemination (AI) of frozen–thawed semen at zoological parks
raffinose) to the cells or tissues before cooling;
around the world (Monfort et al. 1993). For example, AI using
(2) seeding the samples a few degrees below the freezing
frozen–thawed spermatozoa has been used with success in the
point of the cell suspension and cooling the cells to a
cheetah, leopard cat and ocelot (Howard 1999). The use of AI
low temperature at rates calculated to minimise intracel-
with thawed spermatozoa also would assist genetic manage-
lular ice formation, then storage at below glass transition
ment through the distribution of genes among geographically
temperatures (in the range of −130 to −196◦ C);
dispersed breeding centres and zoos. Availability of frozen sper-
(3) thawing the cells; and
matozoa would permit international transport of genetic material
(4) removing the CPA from the cells and rehydrating them
without compromising political and legal restrictions. Effective
before utilisation.
sperm cryopreservation could salvage precious genetic material
from wild animals that are captured in the field for various rea- In general, we may expect coupled flows of water and CPA
sons. Application of assisted reproduction technologies (ART) when CPA are added, during freezing, thawing, and when CPA
using frozen–thawed gametes of rare and endangered species are removed from cells. During the cryopreservation process,
will become increasingly important in conservation biology cells are subjected to a series of anisosmotic conditions. Cells
programs (Wildt 2000). are initially subjected to a hyperosmotic environment during the

© CSIRO 2007 10.1071/RD07035 1031-3613/07/060719


720 Reproduction, Fertility and Development A. Dinnyes et al.

addition of the CPA, which is usually at a concentration of around variable among cell types. In some cells warming rate makes
1.0 M or less for sperm cells. They shrink as water is driven out of little or no difference, but in others, rapid or slow warming is
the cells by osmosis then return to their isosmotic volumes as the required, and is often dependent on the rate of cooling (Mazur
permeating CPA enters the cells and water follows to maintain 1984).
chemical potential equilibrium across cell membranes. During Mazur developed a theoretical model to study how cells lose
freezing the cells dehydrate and shrink as the water in the cell intracellular water during the cryopreservation process at a spe-
suspension gradually becomes frozen below the freezing point cific cooling rate (Mazur 1963). Over the years others have added
of the more and more concentrated solution. The cells remain to this model to develop more refined methods of mathematically
shrunken during storage, then return to their isosmotic volume predicting optimal conditions for cell cryopreservation process-
again upon thawing. Finally, the cells are subjected to potentially ing, and a convergence of theoretical and empirical approaches
lethal swelling upon CPA dilution and removal. These shrinkage has begun. With recent progress in understanding mechanisms of
and swelling events are capable of causing damage or even cell injury to cells during cryopreservation, physical modelling using
death (Mazur and Schneider 1986). mathematical formulations has been developed to simulate a
During the cooling process, if cells are cooled too rapidly, cell’s response to environmental change during the cryopreserva-
water does not exit the cells fast enough to maintain equilibrium. tion process and to predict optimal cryopreservation conditions
The cells become increasingly supercooled and eventually freeze (Toner et al. 1990; Pitt et al. 1992; Karlsson et al. 1994; Liu et al.
intracellularly (Mazur 1990). In most cases, intracellular ice for- 2000). More advanced knowledge on the biophysical character-
mation (IIF) is lethal for cells (Muldrew and McGann 1994). istics of mammalian spermatozoa have been used to optimise
If cooling is relatively slow, the cells will lose water rapidly cryopreservation protocols (Gao et al. 1995; Gilmore et al.
enough to concentrate the intracellular solutes sufficiently to 1997).
eliminate supercooling. Although rates low enough to prevent Recently, new cryopreservation methods have emerged such
internal freezing injury are necessary for high survival, freezing as freeze drying and vitrification.
at slow rates over a long duration can cause ‘solution effects’
injury resulting from the extreme concentration of extracellu-
lar and intracellular solutes that may decrease survival as well Freeze-dried spermatozoa
(Critser et al. 2002), probably due to the effects of the solutes on Cryopreservation of spermatozoa, oocytes and embryos, as well
the cellular membrane or through osmotic dehydration. There- as somatic cells or cell lines for cloning by nuclear transfer, are all
fore, a cooling rate either ‘too high’ or ‘too low’ can kill cells, options for the long-term storage of unique genotypes and endan-
although the mechanisms underlying cell damage are different. gered species. Sperm cryopreservation and storage currently
Typically, cells that survive cryopreservation respond to cooling requires liquid nitrogen or ultra-low refrigeration-based meth-
rates in an inverted ‘U’-shaped pattern, hence there is a cool- ods for long- or short-term storage, with routine maintenance
ing rate for maximum survival. This rate, however, can vary and extensive space requirements.
greatly depending on the cell type (Mazur 1963). Current sperm Besides the current methods available for cryopreservation
cryopreservation protocols commonly utilise cooling rates of of spermatozoa by equilibrium freezing (see previous section),
10–100◦ C min−1 . recently, freeze drying has also been used to preserve mammalian
A cell that has survived cooling to low subzero temperatures spermatozoa, serving as an alternate method of cryopreserva-
still faces challenges during warming and thawing, which can tion (Wakayama and Yanagimachi 1998; Kusakabe et al. 2001;
exert effects on survival comparable to those of cooling (Mazur Kaneko et al. 2003a, 2003b; Ward et al. 2003; Liu et al.
et al. 1984). These effects depend on whether the prior rate of 2004; Poleo et al. 2005). Because freeze-dried spermatozoa can
cooling has induced intracellular freezing or cell dehydration. In be stored at an ambient or refrigerated temperature, the costs
the former case, if the cells are not destroyed through lysis from required for the maintenance and shipping of spermatozoa would
IIF, they may end up with an accumulation of smaller, thermody- be greatly reduced.
namically unstable ice crystals that may subsequently undergo Freeze drying is a procedure designed for easy preserva-
recrystallisation, forming bigger ice crystals that rupture the cell tion and transportation of biological materials (such as viruses,
membrane thus leading to fatal damage. Rapid thawing can res- bacteria, yeasts and fungi), pharmaceuticals and other delicate,
cue many cells, possibly by preventing the harmful growth of solvent-impregnated materials. It is achieved through a process
small intracellular ice crystals by recrystallisation. in which frozen material is dried by the sublimation of ice.
The most important characteristics of CPA solutions are that The freeze drying procedures for spermatozoa described ear-
they are, for the most part, readily able to permeate spermato- lier allow storage of the samples in sealed ampoules at 4◦ C or
zoa and are relatively non-toxic in concentrations of 1 M or less. room temperature (Kusakabe et al. 2001; Kaneko et al. 2003b;
The primary basis for their protective action is that they lower Ward et al. 2003; Kaneko and Nakagata 2005).
the concentration of electrolytes during freezing and therefore The drying of cells usually leads to damage of cellular
decrease the extent of osmotic shrinkage at a given temperature. membranes and proteins, which often results in cell death.
The extent of protection depends primarily on the molar ratio The removal of intracellular water causes changes in molecu-
of the CPA to endogenous solutes inside and outside the cells, lar interactions. When cell membranes are dehydrated, the water
and the general protective mechanism of action is colligative. molecules that help to maintain the spacing between the polar
If cells are cooled slowly enough to avoid intracellular freez- phospholipid head groups are removed. The result is that the lipid
ing through dehydration, the response to warming rate is highly fatty acid chains and polar head groups come closer to each other
Novel gamete storage Reproduction, Fertility and Development 721

and the membrane collapses; an increase in the membrane phase deleterious effects of exposure to CPA can be avoided by opti-
transition temperature (Tm) occurs as the membrane changes mising regimens for their addition and removal (Gao et al. 1995;
from a biologically active fluid phase to a gel phase (Crowe Katkov et al. 1998), as described for human and animal sperma-
et al. 1992, 1997). Subsequently, as water is added back during tozoa. To minimise osmotic and toxic effects associated with
rehydration, the membrane undergoes another phase transition, concentrated CPA, many researchers are exploring vitrification
resulting in leakage of soluble cell contents through each mem- methods that would require lower CPA concentrations. Toxic-
brane (Leslie et al. 1994; Crowe et al. 1997). In the conventional ity can be reduced by combining two CPA or adding precooled
sense, freeze-dried spermatozoa are dead. They are non-motile solutions. However, it is possible to lower the critical perme-
and unable to fertilise either in vivo or in vitro, but they are able able CPA concentration by adding non-permeable CPA (Fahy
to produce live offspring when injected into oocytes by the pro- et al. 1984; Fahy 1986a, 1986b). Additives such as disaccharides
cedure of intracytoplasmic sperm injection (ICSI). Apparently, (e.g. sucrose or trehalose) or high molecular weight molecules
the procedure for this level of freeze drying does not seem to (e.g. Ficoll, poly vinyl alcohol or poly vinyl pyrrolidone) can
affect sperm nuclear DNA integrity. significantly reduce the amount of permeable CPA required.
Recently, a significant effort has been directed at preserving However, these methods also have their limitations. Regardless
spermatozoa in the dry state. In 1992, Katayose and colleagues of the mechanism of damage, in the majority of species, sperma-
demonstrated that hamster and human spermatozoa can be used tozoa cannot tolerate the high concentrations of cryoprotectants
following storage in a dehydrated state for 12 months at 4◦ C used conventionally in vitrification.
(Katayose et al. 1992). They used the ICSI procedure and The obvious alternative is thus to increase cooling and warm-
injected the reconstituted freeze-dried spermatozoa into hamster ing rates to meet the requirement of vitrification. To achieve
oocytes, and then examined the formation of pronuclei. Later, even higher cooling rates, the volume of vitrification solution
the same group reported 85 and 90% pronuclear formation for should be minimised by using specially designed cell suspension
hamsters and humans, respectively (Hoshi et al. 1994). Live mice containers such as open-pulled straws (OPS) (Vajta et al. 1997,
have been produced by ICSI (Kimura andYanagimachi 1995). To 1998), electron microscope copper grids (Martino et al. 1996),
date, embryonic development after ICSI with freeze-dried sperm cryoloops (Lane et al. 1999), cryotops (Kuwayama and Kato
heads has been reported in large domestic animals, including cat- 2000), gel-loading tips (Tominaga and Hamada 2001) and other
tle (Keskintepe et al. 2002) and pigs (Kwon et al. 2004), and live similar devices, or even without any containers in minimum-size
offspring in mice (Wakayama and Yanagimachi 1998; Kusakabe drops (Arav and Zeron 1997), in microdrops (Papis et al. 2000)
et al. 2001; Kaneko et al. 2003a, 2003b; Ward et al. 2003), rab- or by solid-surface vitrification (SSV) (Dinnyés et al. 2000).
bits (Liu et al. 2004), rat (Hirabayashi et al. 2005) and fish (Poleo These methods have been used to achieve better results for the
et al. 2005) have been produced. vitrification of embryos in species that are particularly suscep-
tible to cryodamage and for human and animal oocytes (Chen
et al. 2000; Yoon et al. 2003).
Vitrification as an alternative to conventional freezing Although the application of vitrification started with embryos
Cryopreservation without ice formation (vitrification) could be and oocytes, recently Nawroth et al. (2002) carried out a chal-
beneficial in comparison to freezing methods because it does not lenging experiment with sperm vitrification. They used a copper
need any expensive equipment and takes only a few seconds for loop of 0.5-cm diameter, with 20 µL liquid attached. They esti-
cooling and warming. Vitrification is considered an attractive mated the thickness of the liquid film to be as small as 0.7 mm
alternative to conventional freezing and has been used success- and the speed of cooling as high as 720 000 K min−1 . Using 10%
fully for the cryopreservation of spermatozoa, embryos, oocytes, serum albumin in their standard sperm holding medium, they
stem cells and organs. believed that it was possible to vitrify human spermatozoa in the
Vitrification is a process of converting a material into a glass- absence of cryoprotectants, and they succeeded. They vitrified
like amorphous solid that is free of any crystalline structure, swim-up-prepared spermatozoa and compared their results with
achieved by rapid cooling and mixing with high concentrations the conventional glycerol freezing method. Immediately after
of CPA. In general, the rate of cooling or warming and the con- thawing, swim-up-prepared spermatozoa frozen conventionally
centration of cryoprotectants required to achieve vitrification are with cryoprotectants showed 38% motility compared with 49%
inversely related. In other words, the faster cooling and warm- after vitrification without cryoprotectants. The numbers of mor-
ing is undertaken, the lower the critical solute concentration phologically normal spermatozoa recovered after conventionally
necessary to obtain ice-free vitrification. frozen swim-up-prepared spermatozoa with cryoprotectant was
In 1985, Rall and Fahy successfully vitrified embryos using not different from the vitrified ones (27 v. 26%, respectively)
high CPA concentrations and a relatively low speed of cooling (Nawroth et al. 2002; Isachenko 2003).
and warming (Rall and Fahy 1985). This breakthrough opened Two basic variables of success or failure of vitrification are
the avenue for the practical use of vitrification. A typical vit- the size of the cells and the relative concentration of soluble
rification requires a high concentration of cryoprotectants in macromolecules within the cells. First, the shape and size of
the medium (30–50% compared with 5–10% for slow freezing) the sperm head could define the cryosensitivity of the cell.
at a cooling rate easily achievable for conventional labora- Comparative studies (Nauk 1991) in mammals (boar, bull, ram,
tory conditions and set ups. Such high CPA concentrations rabbit, cat, dog, horse and human) have shown a negative corre-
can be damaging to cells, causing both biochemical alterations lation between the size of the sperm head and cryostability. The
and lethal osmotic injury (Fahy et al. 1984). Some of the results show that the human spermatozoon is the smallest with
722 Reproduction, Fertility and Development A. Dinnyes et al.

Table 1. Comparison of the three approaches for cryopreservation of oocytes


CPA, cryoprotective agent; NA, not applicable

Parameters Cryopreservation techniques


Freezing In-straw vitrification Ultrarapid vitrification

Oocyte storage container Straw Straw Cryovial, straws, special


devices or no container
CPA concentration 1.3–1.5 M 5.5–7.5 M 3.5–5.5 M
Time in CPA (concentration NA 2–5 min 2–5 min
for equilibration)
Time in CPA (concentration 15–20 min ≥1 min ≥10 s
for cooling)
Time required for cooling 90–120 min 2–3 min <0.1 s
Ice crystal formation Yes No No
Osmotic injury Low risk High risk High risk
Toxic injury Low risk High risk High risk
Chilling injury High risk Low risk Low risk
Warming rate Low to moderate Moderate to high High
Cost High Low Low
Commercial applications Extensive Limited Limited

the highest cryostability. Second, spermatozoa naturally con- (Gordts et al. 1990; Kuleshova et al. 1999; Chung et al. 2000)
tain high concentrations of proteins, which helps vitrification. and equine (Tharasanit et al. 2006).
Usually, sperm cells have a very high osmotically inactive vol- The development of female gamete cryopreservation tech-
ume (∼60%), hence osmotically inactive water is also higher in niques in dogs and cats is important both for these companion
spermatozoa and is bound to several macromolecular structures animal species and for the conservation of related endangered
such as DNA, histones and hyaluronidase. Consequently, this species. Research is still in progress in the study of dog oocyte
would enhance both the viscosity and glass transition tempera- maturation, which is still at a much lower rate than most domestic
ture (Tg) of the intracellular cytosol for spermatozoa, whereas species. Problems are due to dissimilarities of the reproductive
for embryos the probability of lethal ice formation during cool- physiology of the dog compared with other species and the lack
ing and warming would be several orders of magnitude higher. of precise information concerning the oviductal environment,
Extensive compartmentalisation of intracellular compounds may in which oocyte maturation, fertilisation and early embryonic
also contribute to the successful survival of spermatozoa. It is development take place (Yamada et al. 1993). In cat, success-
this combination of the unique characteristics of spermatozoa ful in vitro embryo production has been achieved with oocytes
and a high speed of cooling and warming that makes it possible matured in vitro, and kittens were born after transfer (Luvoni
to successfully vitrify human spermatozoa without CPA. 2006). Although sperm cells have been frozen successfully in
dogs and cats, cryopreservation of oocytes in dogs remains elu-
Cryopreservation methods for oocytes sive (Luvoni 2000). Cat oocyte cryopreservation has had some
Successful cryopreservation of female gametes is a viable tool success (Comizzoli et al. 2004). More research on cryostorage
for preserving the female genome and to assist in rescuing endan- of female gametes in dogs and cats is needed, together with refin-
gered species. Subsequently, development of cryopreservation ing techniques for oocytes maturation and fertilisation in vitro
techniques for preserving gametes of wild animals relies greatly to allow for establishment of a cryogene bank.
on the results obtained in livestock and laboratory species. How- Recent efforts have been devoted to improve the survival of
ever, they are not always suitable models for conserving wild frozen or vitrified oocytes by developing cryopreservation tech-
species. To date, research has demonstrated remarkable differ- niques. Currently, there are three strategies for female gamete
ences among species. It is clear from the studies that optimal storage: freezing, traditional vitrification (vitrification in straw)
cryopreservation methods tend to be species specific, due largely and ultra-rapid vitrification (Table 1). Slow freezing has the
to variations in gamete size, permeability and sensitivity to advantage of using low concentrations of CPA, which can reduce
cryoprotectant and cooling or warming rates. chemical toxicity and osmotic shock. Vitrification is a rapid
Cryopreservation of oocytes is one of the greatest challenges method, which decreases chilling injury; however, it requires
facing ART today. However, survival rates of cryopreserved high concentrations of CPA (Rall and Fahy 1985). Recent emerg-
oocytes are lower, with poor developmental competence com- ing ultra-rapid vitrification techniques have stimulated more
pared with their fresh counterparts in hamster (Barnett et al. research on the cryopreservation of female gametes, including
1996), rabbit (Carroll et al. 1989), pig (Rojas et al. 2004; Somfai various endangered species and indigenous livestock breeds, as
et al. 2006; Wu et al. 2006; Gupta et al. 2007), mouse (Rall and there has been greater success in the cryopreservation of female
Fahy 1985; Glenister et al. 1987), cow (Vajta et al. 1998; Dinnyés gametes using methods such as OPS, SSV, cryoloop, cryotop
et al. 2000; Lane and Gardner 2001; Schmidt et al. 2004), human and Vitmaster (Arav et al. 2002).
Novel gamete storage Reproduction, Fertility and Development 723

Advantages of gamete cryopreservation of spermatozoa have been carried out empirically, resulting in
Sperm cryopreservation has been used extensively in human many improvements, including the development of semen exten-
reproductive medicine and sperm banking (Critser and Linden ders such as egg yolk and skim milk, and cholesterol-loaded
1995). It allows donor semen to be screened for sexually trans- cyclodextrins (Watson 1995; Moce and Graham 2006). However,
mitted diseases such as HIV and hepatitis before clinical use considerable loss of viability is still observed. Attempts to adapt
(Mascola and Guinan 1986), whereas cryopreservation of male effective methods from one species to another has resulted in lim-
gametes at various developmental stages (i.e. spermatozoa, elon- ited success (Kumar et al. 2003). In some species, methods with
gated or round spermatids) can be used to provide more advanced efficiency acceptable for the breeders are yet to be determined,
treatment options in the case of spontaneous or iatrogenic infer- whereas in other species, such as mouse, methods that seem
tility (Hallak et al. 2000). Mouse spermatozoa and embryo cry- adequate for some strains fail in others (Critser and Mobraaten
opreservation are major tools for maintaining specific transgenic 2000). Spermatozoa of carnivores cannot be cryopreserved effi-
or knockout mouse strains and lines that are crucially important ciently using standard livestock methodologies (Farstad 2000).
to biomedical research (Critser and Mobraaten 2000). Modern These results indicate that fundamental cryobiological proper-
ART, especially AI and to a lesser extent in vitro fertilisation ties of spermatozoa and examination of the exact nature of sperm
(IVF) and ICSI supported by gamete and embryo cryopreserva- cryoinjury must include a species-specific design in order to
tion, have revolutionised domestic animal breeding programs. develop appropriate cryopreservation protocols (Holt 2000).
These technologies have accelerated the progress of genetic Various aspects of sperm cryopreservation, such as chemical
improvement and have enabled distribution of germplasm world- composition of extenders and their effects on the sperm plasma
wide (Watson 1990). Gradually, these technologies are being membrane, osmotic tolerance limits, hydraulic conductivity and
used in conservation efforts for endangered species and captive CPA permeability have also been studied (Gilmore et al. 1997,
breeding programs (Johnston and Lacy 1995). 1999, 2000).Apart from the damage associated with freezing and
Oocyte cryopreservation in mammalian and endangered thawing that has been discussed in previous sections, oxidation
species has a series of potential benefits and applications aimed of sperm membrane lipids (Brouwers et al. 2005) and damage to
at maintaining gene pool diversity, regenerating populations after the selective permeability mechanisms of the membrane are the
disease outbreaks, rescuing rare or endangered species, provid- most damaging factors in freezing. In addition, the most sensitive
ing a source of genetic material for research purposes, supplying part of the sperm cells are their special acrosomal membranes
germplasm for new line and breed development, and global (Friend and Rudolf 1974; Grondahl et al. 1994). Freeze-fracture
germplasm trading. Therefore, successful cryopreservation of studies of membranes before, during and after cooling show clear
oocytes provides insurance for existing genetic diversity within evidence of phase separation, which is only partially reversed
a species. after warming (Holt and North 1984; de Leeuw et al. 1990).
Many detrimental effects of freezing and thawing on sperm
capacitation (Watson 1995) and DNA denaturation (Gillan and
Complications of gamete cryopreservation
Maxwell 1999) have been reported. Studies suggest that sper-
Complications of cryopreserving spermatozoa matozoa do indeed have different cryobiological properties and
Protocols for cryopreservation of spermatozoa from various various degrees of sensitivity to cold shock, manipulation (e.g.
species have been established, including those for stallion pipetting, centrifugation) and osmotic tolerance in every species
(Graham 1996; Ecot et al. 2000), boar (Johnson et al. 2000), (Katkov and Mazur 1999; Phelps et al. 1999).
goat (Leboeuf et al. 2000), ram (Salamon and Maxwell 2000) Dessicated (lyopreserved) spermatozoa would provide long-
and dog (Rodriguez-Martinez et al. 1993). Although these proto- term storage without the need for expensive and burdensome
cols have integrated the optimised freezing procedures, they do cryogenic conditions, and it may provide a new perspective in
not achieve much more than 50% motility after thawing (Watson genome banking and become part of developing ART. In mouse,
2000; Vishwanath and Shannon 2000), whereas the fertilising Wakayama andYanagimachi (1998) were the first to produce live
ability of the spermatozoa is reduced approximately seven-fold offspring after ICSI from frozen-dried spermatozoa. However,
(Watson 2000; Sullivan 2004). the procedure is still in its experimental stages, and requires
There is a vast diversity in the cryobiological response of further investigation for both efficiency and safety. Some of
spermatozoa among different mammalian species and among the previous studies have examined the cytogenic status of
different individuals within a species (Holt 2000). According to zygotes (fertilised by ICSI), such as chromatin decondensation,
Woods et al. (2004) the unavoidable variations in the success of pronucleus formation, sperm aster formation and chromosome
freezing are due to essential biological variability. Establishing abnormalities, in non-human primates, rabbit, as well as ham-
cryopreservation procedures for a specific cell requires optimi- ster oocytes injected with human spermatozoa (Goud et al. 1998;
sation for that cell type within a species, and among individuals Hewitson et al. 2000; Terada et al. 2000).
in that species. Cryosurvival requires that freezing and thawing One potential difficulty in the application of lyopreserved
are carried out within specific biophysical and biological limits spermatozoa is aberrant decondensation of sperm heads and
defined by the underlying fundamental cryobiological charac- oocyte activation. In rats, Hirabayashi et al. (2005) reported that
teristics of each species’ spermatozoa. In the 1950s, soon after viable offspring can be obtained by intracytoplasmic injection
the positive effect of glycerol was discovered, a great increase in of freeze-dried and rehydrated spermatozoa and that the ultra-
the use of AI with frozen spermatozoa for dairy and beef cattle sonic treatment of spermatozoa is effective in increasing the
followed. Since then, attempts to optimise the cryopreservation production of ICSI-derived offspring. They had established
724 Reproduction, Fertility and Development A. Dinnyes et al.

protocols for microinsemination, including ICSI, elongating (a) (b)


PB
spermatid injection and round spermatid injection. The effi-
ZP
ciency of bovine ICSI is limited because of the necessity for
additional oocyte activation after the ICSI procedure. Studies MT
Nuclear
with activation protocols have demonstrated that the develop- Spindle
membrane
mental competence of bovine embryos could be improved by ZP
treatment with ionomycin followed by 6-dimethylaminopurine
after ICSI (Rho et al. 1998; Keskintepe et al. 2002). GV with DNA
Storing spermatozoa in liquid nitrogen can preserve their bio- MII oocyte
GV oocyte
logical function for many years (Mazur 1984); in bovine, good
fertilisation rates have been achieved using even 37-year-old cry- Fig. 1. The typical structure of immature and mature oocytes. (a) The
opreserved bovine spermatozoa (Leibo et al. 1994). However, immature oocyte at the germinal vesicle (GV) stage is full size but its
long-term effects of dessication on sperm genetic stability dur- chromatin remains at the diplotene stage of prophase one with a nuclear
ing storage in various media is unknown. The long-term effects membrane and zona pellucida (ZP), and it does not possess a spindle appa-
of desiccated spermatozoa are estimated theoretically by acceler- ratus. (b) In matured oocyte (MII stage), the oocyte has undergone nuclear
ated degradation kinetics. Calculated extrapolation of Arrhenius and cytoplasmic maturation, the first polar body (PB) has been expelled and
plots indicates that mouse development rate to the blastocyst the chromosomes are condensed and arranged on the delicate MII spindle.
stage would not decline significantly for 100 years of storage The meiotic spindles consist of microtubules (MT). Microtubules start from
at −80◦ C. The chromosome integrity could be maintained by microtubular organising centres at both poles and anchor chromosomes at
the kinetochores, forming a barrel shape. The chromosomes align at the
adding calcium-chelating agents during freeze drying (Kawase equatorial plane of the meiotic spindles.
et al. 2005).
Indeed, it was found that structural chromosomal abnor-
malities were significantly higher in mouse oocytes injected
with spermatozoa freeze-dried in Chatot, Ziomet and Bavister limited, and the current live birth rate is low (Rutherford and
medium. However, chromosome integrity during freeze dry- Gosden 1999). Although female gametes have been cryopre-
ing can be maintained using Tris-HCl-buffered solution with served using various protocols, the fertilisation and in vitro
high concentrations of a calcium-chelating agent (Kaneko and development rates achieved are much lower than those obtained
Nakagata 2005). with fresh oocytes.
Vitrification of human spermatozoa in the absence of conven- Difficulties in the cryopreservation of mammalian oocytes
tional cryoprotectants is feasible. The DNA integrity of vitrified are due to their complex structure (Massip et al. 1987; Friedler
spermatozoa is comparable with that shown by standard slow- et al. 1988; Rall 1992; Arav et al. 1993; Smorag and Gajda
frozen–thawed spermatozoa, yet the method is quick and simple 1994; Kuwayama and Kato 2000). Oocytes are among the largest
and does not require special cryobiological equipment. This mammalian cells, with a low surface-to-volume ratio. Further-
technology has great potential for application in companion ani- more, the oocyte’s cytoskeleton structure can be sensitive to
mals, non-domestic and endangered species (CANDES) in field cooling. At the time of ovulation, in most species oocytes are
conditions, although so far only human spermatozoa have been in metaphase stage of the second division (MII). The dichro-
vitrified successfully. Other shortcomings could be the limita- matid chromosomes aligned on the equatorial axis are bound
tions of the amount of sample held by each cryoloop and sperm to the microtubules in the meiotic spindle (Fig. 1). This stage
prewashing. Considering that many CANDES have low sperm can be problematic for cryopreservation (Bernard and Fuller
concentrations in their collected ejaculates, it might be imprac- 1996; Wood et al. 2001) with temperature and CPA sensitiv-
tical to use many cryoloops to vitrify one AI dosage; thus, the ity of the spindle and microtubules (Newton 1998; Arav et al.
method might be more suitable when combined with ICSI. 2002). In addition, the lipid content of the membranes can change
In situ cryostorage of spermatozoa by freezing testis, or even CPA and water permeability. The large size of oocytes with
whole animals, could be an interesting option and potentially consequent low surface-to-volume ratios make it more diffi-
very advantageous in CANDES. Recently Ogonuki et al. (2006) cult for water and cryoprotectants to enter or leave the cells
reported high success rates of ICSI from frozen testis and epi- across plasma membranes (Leibo and Songsasen 2002), thus
didymidis. Surprisingly, progeny were generated by ICSI with increasing the chances of ice crystal formation and CPA tox-
spermatozoa recovered from whole mouse stored at −20◦ C for icity. Low temperature causes depolymerisation of the spindle
15 years. This not only shows the resistance of sperm DNA and microtubules. As a result, fertilisation may not take place
to damage, but would offer a very practical option for stor- because normal separation of chromatids might be prevented,
ing smaller CANDES cadavers or reproductive organs under resulting in aneuploidy following extrusion of the second polar
conditions available even in remote areas and non-specialised body. Zona pellucida hardening due to premature cortical gran-
facilities. ule release can be triggered by some CPA and eventually can
prevent normal fertilisation.
Collection of oocytes from secondary follicles of ovaries
Complications of cryopreserving female gametes results in germinal vesicle (GV)-stage oocytes. There have been
Despite the extensive research conducted during the past 20 theoretical grounds for favouring GV-stage oocyte cryopreserva-
years, progress in the cryopreservation of oocytes remains tion. Before GV breakdown, chromosomes are still surrounded
Novel gamete storage Reproduction, Fertility and Development 725

by a nuclear membrane (Wood et al. 2001), making them poten- Cryostorage of ovarian preantral follicles would also pro-
tially more cryotolerant (Agca et al. 2000). However, after vide a source of viable oocytes that could be used to propagate
cryopreservation at this stage, there is a need for GV-stage endangered or other species (Van den Hurk et al. 2000; Biron-
oocytes to mature efficiently in vitro up to the stage where nor- Shental et al. 2004). However, in vitro growth of early follicles
mal fertilisation can be achieved. Moreover, cryopreservation is still very inefficient.
procedures have been reported to have deleterious influences
on chromatin and other cytoplasmic organelles of GV oocytes.
In some cases in vitro maturation following cryopreservation Packaging and biocontainment
of GV oocytes led to higher incidences of chromosomal and Usually, the cryopreservation of gametes of laboratory and
spindle abnormalities and adversely affected postfertilisation domestic animals is conducted in a laboratory where sophis-
development (Van Blerkom and Davis 1994; Son et al. 1996; ticated equipment is available and environmental conditions
Park et al. 1997). The present efficiency and reliability of using and the risk of contamination are controllable. There are issues
cryopreserved oocytes for generating offspring is still much of packaging and biocontainment that require more serious
lower compared with cryopreserved embryos or spermatozoa. consideration for CANDES than for laboratory and domestic
Cryopreservation of oocytes of avian and fish species is not yet species. From a cryobiological point of view, the packaging
successful, largely because of the large size, high lipid content would affect the cooling and warming rates due to different
and polar organisation (vegetal and animal pole) of bird and dimensions and materials used in various packaging systems
fish oocytes (Douard et al. 2005). Fish eggs are sensitive to and appropriate adjustments of parameters in cryopreserva-
osmotic changes, which can result in spontaneous activation. tion protocols are usually needed to compensate for these
However, recent reports on cryopreservation of fish primordial variables.
germ cells followed by their transfer into the abdominal cav- Basic choices of packaging include cryovials or straws. Cry-
ity resulted in offspring (Kobayashi et al. 2007), demonstrating ovials can hold a larger volume of sample (beneficial for sperm
that success can be achieved with such an approach. If inter- preservation); however, cooling rates and actual temperatures in
species transfer of fish primordial germ cells works, this would the vials vary during cooling and warming. Moreover, such vials
help endangered species too. However, in most cases such proce- are only safe to store in the gas phase of nitrogen tanks, as storage
dures would be complicated to carry out. Similarly, although the in liquid nitrogen (although a common practice) usually results
current very inefficient approach for cryopreservation of avian in leakage of the liquid nitrogen into the vials. This can result in
genetics occurs via germinal disk stem cell isolation, cryopreser- cross contamination between vials, and can even blow the vials
vation then introduction of the stem cells into a developing egg up during warming.
would be possible. For example, chimeric chicken and ducks Plastic straws (usually 0.25- and 0.5-mL volumes) are more
have been produced with this method using cryopreserved ger- homogeneous during cooling and warming, with higher cool-
minal cells, and this technology is under development owing ing and warming rates achievable. Powder or metal ball sealings
to its relevance for poultry biotechnological applications (Kino are not fully leak proof, and regular straws are easy to break
et al. 1997). at cryogenic temperatures. Heat sealing can prevent leakage,
and high-security straws (CryoBioSystem, Paris) are especially
suitable for leakage-proof storage and are not fragile at low tem-
Storage of ovarian tissue perature (Mortimer 2004). These samples can be stored without
Considering the complexity of cryopreserving female gametes, any concerns in liquid nitrogen.
ovarian tissue cryostorage may be an alternative solution. Preser- Packaging can affect the effectiveness of biocontainment,
vation of female gametes may be best achieved by storing pieces which is particularly important for CANDES due to the high
of ovarian tissue that contain numerous small follicles. Cryop- risk of contamination and cross-contamination in the field
reservation of ovarian tissue has been reported to be possible conditions. Precautionary steps can reduce risk and damage
in humans, mice, sheep, rats, marmosets, elephants, wombats, substantially, such as disinfecting the outside of the packaging,
tammar wallabies and rabbits (see review by Amorim et al. sterilising the cryopreservation equipment and dividing storage
2003). locations (Mortimer 2004). However, some novel technologies,
Early-stage oocytes in primordial follicles are smaller, posses such as the vitrification of human spermatozoa or oocytes in
fewer organelles and have no zona pellucida or cortical granules; minimum volume systems (OPS, cryoloop, droplets, pellets),
hence, they are potentially easier to cryopreserve (Oktay et al. require direct exposure of samples to liquid nitrogen in order
1998; Paynter 2000). In addition, xenografting of the ovaries of to achieve sufficiently rapid cooling rates, and this usually pre-
3-week-old mice into rats was successful in a generation of pups cludes effective biocontainment. There are efforts to overcome
(Shaw et al. 2000). Interspecies transplantation of ovaries from these problems (e.g. OPS mini-straws packed into 0.5-mL sealed
large mammals, including humans, monkeys, cows, pigs, dogs straws for storage). Also, the SSV method avoids direct contact
and marsupials, to recipient mice has resulted in the develop- with liquid nitrogen.
ment of antral follicles and even matured oocytes (see review During the design of cryopreservation protocols to pre-
by Katska-Ksiazkiewicz et al. 2006). This technique might also serve CANDES, strategic planning must include experimental
be used in wildlife conservation, through the formation of gene conditions as well as risk assessment of contamination and cross-
banks to maintain gene pool diversity in captive breeding (Wildt contamination depending on the choice of packaging, storage
2000; Amorim et al. 2003). and cryogenic equipment.
726 Reproduction, Fertility and Development A. Dinnyes et al.

Conclusion CPA, time of exposure, resistance to osmotic changes, minimal


In the last 50 years, the need for breeding management and critical volume, cell size and morphology greatly influence the
genetic improvement for domestic livestock industries, more occurrence of freezing injury. Taking into account these vari-
advanced treatments for human infertility, efficient preservation ables has laid the foundation for developing routine and effective
of laboratory animal species, and better conservation of bio- cryopreservation protocols for human, bull and stallion sper-
diversity for endangered species have motivated scientists and matozoa. Accumulated knowledge in cryobiology and cellular
breeders to utilise empirical and theoretical research approaches characteristics made it possible to rationally design cooling pro-
in developing efficient cryopreservation procedures for a broad cedures using computer simulations (Karlsson et al. 1994; Liu
range of species. Systematic research on conventional freezing et al. 2000).
methods have lead to improved CPA use, cooling and warm- Simulations in the development of cryopreservation meth-
ing procedures and avoidance of damaging ice formation for ods for CANDES would be highly beneficial as they would save
humans and laboratory and domestic animals. Evidently, these limited resources. In order to achieve this, further fundamental
techniques have been modified and adapted for preserving com- research is required to elucidate the mechanisms of damage dur-
panion, non-domestic and endangered species. Many CANDES ing cryopreservation, and to develop predictive models for these
spermatozoa have been cryopreserved successfully (Blanco et al. processes. Likewise, measurements of the biophysical proper-
2000; Robeck and O’Brien 2004; Si et al. 2004), although in ties of CANDES spermatozoa should be developed in order to
some species there are still major difficulties (Pukazhenthi et al. obtain necessary model parameters.
2006b). One of the promising attempts for CANDES spermatozoa
With the progress of overall ART, new preservation technolo- was made by Blanco et al. (2000) investigating potential factors
gies are emerging too. Vitrification of human spermatozoa in of spermatozoa cryosurvival for several avian species including
the absence of conventional CPA has been shown to be feasible; chicken, turkey, golden eagle, Bonelli’s eagle, imperial eagle
however, more research is needed to demonstrate the long-term and peregrine falcon. These studies focused on sperm toler-
effects of such an approach on fertilisation, fetal development ance toward osmotic stress, concentrations of CPA, equilibration
and progeny in animal models. Dessicated (lyopreserved) sper- times at 4 and 21◦ C, and cryopreservation by rapid or slow
matozoa would provide a beneficial method for genome banking freezing, demonstrating the necessity for species-specific cryop-
and ART. To date, embryonic development after ICSI with reservation protocols for avian species. Thirumala et al. (2003)
freeze-dried sperm heads has been reported in large domestic calculated that canine sperm freezing rates based on membrane
animals, including cattle and pigs, and live offspring in mice, permeability parameters differ significantly from those in other
rat, rabbits and fish have been born. mammalian species. The optimal rate of freezing for canine
Despite progress in freezing and vitrification methods, cry- sperm cells was in the order of tens of ◦ C min−1 , in good
opreservation of mammalian oocytes remains a major challenge accordance with rates obtained empirically.
in ART. Matured oocytes have major problems in terms of Vitrification of human spermatozoa in the absence of con-
temperature and CPA sensitivity, and in some species GV ventional cryoprotectants has been proven experimentally to be
oocytes might be easier to obtain and cryopreserve. However, feasible. However, more research is needed to test whether the
in vitro maturation following cryopreservation of GV oocytes technology can be applied to other species, especially CANDES,
needs species-specific studies as incidences of chromosomal and as it would be very advantageous in eliminating the need for
spindle abnormalities that occur during maturation can reduce sophisticated equipment. As discussed in the previous section,
postfertilisation development. Cryopreservation of oocytes of this technology might require better AI and IVF efficiency to
avian and fish species is not yet successful, largely because of compensate for the small sample size cryopreserved.
the large size, high lipid content and polar organisation, although Although the technology of desiccated (lyopreserved) sper-
novel approaches using germ cells offer some alternatives. matozoa as an alternative to cryopreservation is in the experi-
Cryopreserving ovarian preantral follicles or ovarian tissue mental stage, it has been investigated for laboratory and domestic
would provide a great number of developmentally competent animals. It would provide a convenient method of long-term
oocytes for protocols such as IVF, embryo transfer, cloning and storage, transportation, and an effective way of storing sper-
transgenesis (Van den Hurk et al. 2000). matozoa for some laboratory animals (inbred rat and mouse
strains) in which conventional cryopreservation results in very
poor fertility.
Future directions However, further investigations are required to improve its
The challenges in development of optimal cryopreservation tech- efficiency and feasibility for CANDES. This method is rather
nology for CANDES spermatozoa are due to the diversity of inconvenient and inpractical, as the procedures of microinjec-
cryopreservation conditions and availability of resources in dif- tion, oocyte activation, IVF and embryo transfer are essential
ferent species. The challenge is to determine the optimal protocol parts of the whole process of preservation and rederivation and
among an almost infinite number of possibilities (Karlsson and should be used only as a last resort for CANDES.
Toner 2000). To replace traditional empirical methods, such as Cryopreservation is used widely in the fields of human
modifying technical conditions in freezing trials, novel models medicine, biological research, domestic livestock industries and
have been proposed to predict the likelihood of freezing-induced preservation of CANDES. Although seeking effective and effi-
injury or mortality (Muldrew and McGann 1990, 1994; Toner cient cryopreservation procedures are goals for each scientist
et al. 1990). Membrane permeability to water and permeating in these areas, different areas might define the ‘effectiveness
Novel gamete storage Reproduction, Fertility and Development 727

and efficacy’ differently due to their scientific and economical losing their reproductive capacity or for rescuing endangered
priority and resources. Progress in both conventional and novel species.
cryopreservation methods would definitely provide new choices
for CANDES researchers to select accordingly.
Historically, methods for female gamete storage were devel- Acknowledgements
oped using an empirical approach and the parameters to assess The work was supported by the European Union (MEDRAT-LSHG-CT-
the success of a given protocol have typically been morphologi- 2005-518240 and MRTN-CT-2006-035468), by the Hungarian Govern-
ment (NKTH/KPI Kozma Ferenc TUDAS-1-2006-0005 project), and a
cal survival rate, fertilisation and blastocyst formation. Although
Hungarian–South African Bilateral Scientific and Technological (TET)
this approach has certainly resulted in improved cryopreserva-
collaborative project.
tion techniques and protocols, it has provided limited informa-
tion regarding the actual physiology of the female gamete and References
complex events such as interactions with CPA, effects of cool-
Agca, Y., Liu, J., Critser, E. S., and Critser, J. K. (2000). Fundamen-
ing and warming rates used and impact on cell function. Gene
tal cryobiology of rat immature and mature oocytes: hydraulic con-
expression changes due to cryopreservation have been reported ductivity in the presence of Me2 SO, Me2 SO permeability, and their
for embryos (Boonkusol et al. 2006; Mamo et al. 2006) but very activation energies. J. Exp. Zool. 286, 523–533. doi:10.1002/(SICI)1097-
little is known about changes in gene expression and protein 010X(20000401)286:5<523::AID-JEZ10>3.0.CO;2-9
profiles due to oocyte cryopreservation. More molecular bio- Amorim, C. A., Goncalves, P. B., and Figueiredo, J. R. (2003). Cryopreser-
logical data might help to model mathematically the oocyte as a vation of oocytes from pre-antral follicles. Hum. Reprod. Update 9,
‘system’ and to make reliable predictions for cryobehaviour and 119–129. doi:10.1093/HUMUPD/DMG014
create fundamentally new avenues for cryopreservation taking Arav, A., and Zeron, Y. (1997). Vitrification of bovine oocytes using modi-
into account long-term biological consequences as well. fied minimum drop size technique (MDS) is effected by the composition
and the concentration of the vitrification solution and by the cooling
As reviewed, there is still much to be done to optimise pro-
conditions. Theriogenology 47, 341. [Abstract] doi:10.1016/S0093-
tocols for successful cryostorage of female gametes, especially
691X(97)82468-5
from CANDES. These gametes would be a valuable resource Arav, A., Shehu, D., and Mattioli, M. (1993). Osmotic and cytotoxic study of
contributing to the preservation of genetic material essential for vitrification of immature bovine oocytes. J. Reprod. Fertil. 99, 353–358.
the maintenance and captive management of endangered species. Arav, A., Yavin, S., Zeron, Y., Natan, D., Dekel, I., and Gacitua, H. (2002).
There are still many obstacles to overcome, such as the improve- New trends in gamete’s cryopreservation. Mol. Cell. Endocrinol. 187,
ment of cryopreservation protocols for both male and female 77–81. doi:10.1016/S0303-7207(01)00700-6
gametes and establishment of good and optimised in vitro mat- Barnett, D. K., Kimura, J., and Bavister, B. D. (1996). Translocation of
uration protocols for post-thawing use. Furthermore, effective active mitochondria during hamster preimplantation embryo devel-
techniques in domestic species are not necessarily applicable to opment studied by confocal laser scanning microscopy. Dev. Dyn.
205, 64–72. doi:10.1002/(SICI)1097-0177(199601)205:1<64::AID-
wild species. Reproductive mechanisms are highly varied among
AJA6>3.0.CO;2-3
animal groups.
Bernard, A., and Fuller, B. J. (1996). Cryopreservation of human oocytes: a
Storage of ovarian tissue and early-stage follicles might review of current problems and perspectives. Hum. Reprod. Update 2,
become an alternative resource for the foundation for cryogene 193–207. doi:10.1093/HUMUPD/2.3.193
banks, and for direct storage of CANDES individuals killed acci- Biron-Shental, T., Fisch, B., Van Den Hurk, R., Felz, C., Feldberg, D.,
dentally or by hunting. However, it is too early to predict when and Abir, R. (2004). Survival of frozen–thawed human ovarian fetal
this technology will become practical. Some of these problems follicles in long-term organ culture. Fertil. Steril. 81, 716–719.
might eventually be overcome by growing cryopreserved folli- doi:10.1016/J.FERTNSTERT.2003.06.034
cles and oocytes to maturity in vitro or in temporary hosts, such Blanco, J. M., Gee, G., Wildt, D. E., and Donoghue, A. M. (2000). Species
as ectopic sites of severe combined immunodeficiency diseased variation in osmotic, cryoprotectant, and cooling rate tolerance in poultry,
eagle, and peregrine falcon spermatozoa. Biol. Reprod. 63, 1164–1171.
mice (Kagawa et al. 2007).
doi:10.1095/BIOLREPROD63.4.1164
Female gamete storage has been done mostly by vitrification
Boonkusol, D., Baji Gal, A., Bodo, S., Gorhony, B., Kitiyanant, Y., and
rather than the slow-freezing method. Future female gamete cry- Dinnyes, A. (2006). Gene expression profiles and in vitro development
opreservation might be based mainly on vitrification (Vajta and following vitrification of pronuclear and 8-cell stage mouse embryos.
Kuwayama 2006), although some of the most efficient vitrifica- Mol. Reprod. Dev. 73, 700–708. doi:10.1002/MRD.20450
tion methods are lacking the possibility for direct transfer and Brouwers, J. F., Silva, P. F., and Gadella, B. M. (2005). New assays for detec-
there are concerns of possible disease transmission through liq- tion and localization of endogenous lipid peroxidation products in living
uid nitrogen during vitrification and storage. This might be a boar sperm after BTS dilution or after freeze-thawing. Theriogenology
particularly important issue in the case of CANDES where full 63, 458–469. doi:10.1016/J.THERIOGENOLOGY.2004.09.046
record of health status is often not available. Another area worth Carroll, J., Warnes, G. M., and Matthews, C. D. (1989). Increase in digyny
explains polyploidy after in-vitro fertilization of frozen–thawed mouse
investigating is stabilising cell membranes with trehalose. The
oocytes. J. Reprod. Fertil. 85, 489–494.
drawback is how to transfer trehalose into the cytoplasm of the
Chen, S.-U., Lien, Y.-R., Chao, K.-H., Lu, H.-F., Ho, H.-N., and Yang, Y.-S.
cell, where it could protect internal membranes as well (Seidel (2000). Cryopreservation of mature human oocytes by vitrification with
2006). ethylene glycol in straws. Fertil. Steril. 74, 804–808. doi:10.1016/S0015-
Oocyte cryostorage is improving, especially in cattle, humans 0282(00)01516-8
and some other species, and one day may replace embryo Chung, H. M., Hong, S. W., Lim, J. M., Lee, S. H., Cha, W. T., Ko, J. J.,
cryopreservation offering an alternative to animals at risk of Han, S. Y., Choi, D. H., and Cha, K. Y. (2000). In vitro blastocyst
728 Reproduction, Fertility and Development A. Dinnyes et al.

formation of human oocytes obtained from unstimulated and stimulated and removal from human spermatozoa. Hum. Reprod. 12, 112–118.
cycles after vitrification at various maturational stages. Fertil. Steril. 73, doi:10.1093/HUMREP/12.1.112
545–551. doi:10.1016/S0015-0282(99)00546-4 Gilmore, J. A., Liu, J., Woods, E. J., Peter, A. T., and Critser, J. K. (2000). Cry-
Comizzoli, P., Wildt, D. E., and Pukazhenthi, B. S. (2004). Effect of 1,2- oprotective agent and temperature effects on human sperm membrane
propanediol versus 1,2-ethanediol on subsequent oocyte maturation, permeabilities: convergence of theoretical and empirical approaches
spindle integrity, fertilization, and embryo development in vitro in the for optimal cryopreservation methods Hum. Reprod. 15, 335–343.
domestic cat. Biol. Reprod. 71, 598–604. doi:10.1095/BIOLREPROD. doi:10.1093/HUMREP/15.2.335
104.027920 Gilmore, J. A., Liu, J., and Critser, J. K. (1999). Osmotic tolerance limits of
Critser, J. K., and Linden, J. V. (1995). Therapeutic insemination by donor. murine spermatozoa in the presence of extender media Cryobiology 39,
A review of its efficacy. Reprod. Med. Rev. 4, 9–17. 353–354.
Critser, J. K., and Mobraaten, L. E. (2000). Cryopreservation of murine Glenister, P. H., Wood, M. J., Kirby, C., and Whittingham, D. G. (1987).
spermatozoa. ILAR J. 41, 197–206. Incidence of chromosome anomalies in first-cleavage mouse embryos
Critser, J. K., Agca, Y., and Woods, E. J. (2002). Cryopreservation of mature obtained from frozen–thawed oocytes fertilized in vitro. Gamete Res.
and immature gametes. In ‘Assisted Reproductive Technology’. (Eds 16, 205–216. doi:10.1002/MRD.1120160303
C. J. DeJonge and C. L. R. Barratt.) pp. 144–166. (Cambridge University Gordts, S., Roziers, P., Campo, R., and Noto, V. (1990). Survival and preg-
Press: Cambridge, UK.) nancy outcome after ultrarapid freezing of human embryos. Fertil. Steril.
Crowe, J. H., Hoekstra, F. A., and Crowe, L. M. (1992). Anhydrobiosis. 53, 469–472.
Ann. Rev. Physiol. 54, 579–599. doi:10.1146/ANNUREV.PH.54.030192. Goud, P. T., Goud, A. P., Rybouchkin, A. V., De Sutter, P., and Dhont, M.
003051 (1998). Chromatin decondensation, pronucleus formation, metaphase
Crowe, J. H., Oliver, A. E., Hoekstra, F. A., and Crowe, L. M. entry and chromosome complements of human spermatozoa after intra-
(1997). Stabilization of dry membranes by mixtures of hydroxyethyl cytoplasmic sperm injection into hamster oocytes. Hum. Reprod. 13,
starch and glucose: the role of vitrification. Cryobiology 35, 20–30. 1336–1345. doi:10.1093/HUMREP/13.5.1336
doi:10.1006/CRYO.1997.2020 Graham, J. K. (1996). Cryopreservation of stallion spermatozoa. Vet. Clin.
de Leeuw, F. E., Chen, H.-C., Colenbrander, B., and Verkleij, A. J. N. Am. Equine Practioneers 12, 131–147.
(1990). Cold induced ultrastructural changes in bull and boar sperm Grondahl, C., Grondahl, M. L., Hyttel, P., and Greve, T. (1994). Acro-
plasma membranes. Cryobiology 27, 171–183. doi:10.1016/0011- somal status in fresh and frozen/thawed stallion spermatozoa evalu-
2240(90)90009-S ated by scanning electron microscopy. Anat. Embryol. 190, 195–200.
Dinnyés, A., Dai, Y., Jiang, S., and Yang, X. (2000). High developmental doi:10.1007/BF00193415
rates of vitrified bovine oocytes following parthenogenetic activation, Gupta, M. K., Uhm, S. J., and Lee, H. T. (2007). Cryopreservation of imma-
in vitro fertilization, and somatic cell nuclear transfer. Biol. Reprod. 63, ture and in vitro matured porcine oocytes by solid surface vitrification.
513–518. doi:10.1095/BIOLREPROD63.2.513 Theriogenology 67, 238–248. doi:10.1016/J.THERIOGENOLOGY.
Douard, V., Hermier, D., Labbe, C., Magistrini, M., and Blesbois, E. 2006.07.015
(2005). Role of seminal plasma in damage to turkey sper- Hallak, J., Mahran, A., Chae, J., and Agarwal, A. J. (2000). The effects
matozoa during in vitro storage. Theriogenology 63, 126–137. of cryopreservation on semen from men with sarcoma or carcinoma.
doi:10.1016/J.THERIOGENOLOGY.2004.03.020 J. Assist. Reprod. Genet. 17, 218–221. doi:10.1023/A:1009443901307
Ecot, P., Vidament, M., de Monarc, A., Perigault, K., Clement, F., and Hewitson, L., Simerly, C., Dominko, T., and Schatten, G. (2000). Cellular
Palmer, E. (2000). Freezing of stallion semen: interactions among and molecular events after in vitro fertilization and intracytoplas-
cooling treatments, semen extenders and stallions. J. Reprod. Fertil. mic sperm injection. Theriogenology 53, 95–104. doi:10.1016/S0093-
56(Suppl.), 141–150. 691X(99)00243-5
Fahy, G. M. (1986a). Cryprotectant toxicity reduction: specific or Hirabayashi, M., Kato, M., Ito, J., and Hochi, S. (2005). Viable rat offspring
nonspecific? Cryobiology 23, 1–13. doi:10.1016/0011-2240(86) derived from oocytes intracytoplasmically injected with freeze-dried
90013-1 sperm heads. Zygote 13, 79–85. doi:10.1017/S096719940500300X
Fahy, G. M. (1986b). Vitrification: a new approach to organ cryop- Holt, W. V. (2000). Fundamental aspects of sperm cryobiology: the impor-
reservation. In ‘Transplantation: Approaches to Graft Rejection’. (Ed. tance of species and individual differences Theriogenology 53, 47–58.
H. T. Meryman.) pp. 305–335. (Alan R. Liss: New York.) doi:10.1016/S0093-691X(99)00239-3
Fahy, G. M., McFarlane, D. R., and Angell, C. (1984). Vitrifica- Holt, W. V., and North, R. D. (1984). Partially irreversible cold-
tion as an approach to cryopreservation. Cryobiology 21, 407–426. induced lipid phase transitions in mammalian sperm plasma mem-
doi:10.1016/0011-2240(84)90079-8 brane domains: freeze-fracture study. J. Exp. Zool. 230, 473–483.
Farstad, W. (2000). Assisted reproductive technology in canid species. doi:10.1002/JEZ.1402300316
Theriogenology 53, 175–186. doi:10.1016/S0093-691X(99)00250-2 Hoshi, K., Yanagida, K., Katayose, H., and Yazawa, H. (1994). Pronuclear
Friedler, S., Giudice, L. C., and Lamb, E. J. (1988). Cryopreservation of formation and cleavage of mammalian eggs after microsurgical injection
embryos and ova. Fertil. Steril. 49, 743–764. of freeze-dried sperm nuclei. Zygote 2, 237–242.
Friend, D. S., and Rudolf, I. (1974). Acrosomal disruption in sperm. Howard, J. G. (1999).Assisted reproduction techniques in carnivores. In ‘Zoo
Freeze-fracture of altered membranes. J. Cell Biol. 63, 466–479. and Wild Animal Medicine IV’. (Eds M. E. Fowler and R. E. Miller.) pp.
doi:10.1083/JCB.63.2.466 449–457. (W. B. Saunders Co.: Philadelphia.)
Gao, D. Y., Liu, J., Liu, C., McGann, L. E., Watson, P. F., Kleinhans, F. W., Isachenko, E. (2003).Vitrification of mammalian spermatozoa in the absence
Mazur, P., Critser, E. S., and Critser, J. K. (1995). Prevention of osmotic of cryoprotectants: from past practical difficulties to present success.
injury to human spermatozoa during addition and removal of glycerol. Reprod. Biomed. Online 6, 191–200.
Hum. Reprod. 10, 1109–1122. Johnston, L. A., and Lacy, R. C. (1995). Genome resource banking for
Gillan, L., and Maxwell, W. M. C. (1999). The functional integrity and fate species conservation: selection of sperm donors. Cryobiology 32, 68–77.
of cryopreserved ram spermatozoa in the female tract. J. Reprod. Fertil. doi:10.1006/CRYO.1995.1006
Suppl. 54, 271–283. Johnson, L. A., Weitze, K. F., Fiser, P., and Maxwell, W. M. C. (2000). Storage
Gilmore, J. A., Liu, J., Gao, D. Y., and Critser, J. K. (1997). Determi- of boar semen. Anim. Reprod. Sci. 62, 143–172. doi:10.1016/S0378-
nation of optimal cryoprotectants and procedures for their addition 4320(00)00157-3
Novel gamete storage Reproduction, Fertility and Development 729

Kagawa, N., Kuwayama, M., Nakata, K., Vajta, G., Silber, S. J., Manabe, N., mouse spermatozoa. Proc. Natl Acad. Sci. USA 98, 13 501–13 506.
and Kato, O. (2007). Production of the first offspring from oocytes doi:10.1073/PNAS.241517598
derived from fresh and cryopreserved pre-antral follicles of adult mice. Kuwayama, M, and Kato, O. (2000). Successful vitrification of human
Reprod. Biomed.Online 14, 693–699. oocytes. Fertil. Steril. 74(Suppl. 3), S49. [Abstract] doi:10.1016/S0015-
Kaneko, T., and Nakagata, N. (2005). Relation between storage temperature 0282(00)00850-5
and fertilizing ability of freeze-dried mouse spermatozoa. Comp. Med. Kwon, I. K., Park, K. E., and Niwa, K. (2004). Activation, pronuclear forma-
55, 140–144. tion, and development in vitro of pig oocytes following intracytoplasmic
Kaneko, T., Whittingham, D. G., Overstreet, J. W., and Yanagimachi, R. injection of freeze-dried spermatozoa. Biol. Reprod. 71, 1430–1436.
(2003a). Tolerance of the mouse sperm nuclei to freeze-drying doi:10.1095/BIOLREPROD.104.031260
depends on their disulfide status. Biol. Reprod. 69, 1859–1862. Lane, M., and Gardner, D. K. (2001). Vitrification of mouse oocytes
doi:10.1095/BIOLREPROD.103.019729 using a nylon loop. Mol. Reprod. Dev. 58, 342–347. doi:10.1002/1098-
Kaneko, T., Whittingham, D. G., and Yanagimachi, R. (2003b). Effect of 2795(200103)58:3<342::AID-MRD13>3.0.CO;2-X
pH value of freeze-drying solution on the chromosome integrity and Lane, M., William, B., Schoolcraft, M. D., and Gardner, D. K. (1999).
developmental ability of mouse spermatozoa. Biol. Reprod. 68, 136–139. Vitrification of mouse and human blastocysts using a novel cryoloop
doi:10.1095/BIOLREPROD.102.008706 container-less technique Fertil. Steril. 72, 1073–1078. doi:10.1016/
Karlsson, J. O. M., and Toner, M. (2000). Cryopreservation. In ‘Principles S0015-0282(99)00418-5
of Tissue Engineering, 2nd edition’. (Eds R. P. Lanza, R. Langer and Leboeuf, B., Restall, B., and Salamon, S. (2000). Production and storage of
J. P. Vacanti.) pp. 293–307. (Academic Press: San Diego.) goat semen for artificial insemination. Anim. Reprod. Sci. 62, 113–141.
Karlsson, J. O., Cravalho, E. G., and Toner, M. (1994). A model of diffusion- doi:10.1016/S0378-4320(00)00156-1
limited ice growth inside biological cells during freezing. J.Appl. Physiol. Leibo, S. P., and Songsasen, N. (2002). Cryopreservation of gametes
75, 4442–4450. doi:10.1063/1.355959 and embryos of non-domestic species. Theriogenology 57, 303–326.
Katayose, H., Matsuda, J., and Yanagimachi, R. (1992). The ability of dehy- doi:10.1016/S0093-691X(01)00673-2
drated hamster and human sperm nuclei to develop into pronuclei. Biol. Leibo, S. P., Semple, M. E., and Kroetsch, T. G. (1994). In vitro
Reprod. 47, 277–284. doi:10.1095/BIOLREPROD47.2.277 fertilization of oocytes by 37-year-old cryopreserved bovine sper-
Katkov, I. I., and Mazur, P. (1999). Factors affecting yield and survival of matozoa. Theriogenology 42, 1257–1262. doi:10.1016/0093-691X(94)
cells when suspensions are subjected to centrifugation. Influence of cen- 90245-E
trifugal acceleration, time of centrifugation, and length of the suspension Leslie, S. B., Teter, S. A., Crowe, L. M., and Crowe, J. H. (1994). Trehalose
column in quasi homogeneous centrifugal fields Cell Biochem. Biophys. lowers membrane phase transitions in dry yeast cells. Biochim. Biophys.
31, 231–245. Acta 1192, 7–13. doi:10.1016/0005-2736(94)90136-8
Katkov, I. I., Katkova, N., Critser, J. K., and Mazur, P. (1998). Mouse sperma- Liu, J., Woods, E., Agca, Y., Critser, E. S., and Critser, J. K. (2000). Cry-
tozoa in high concentrations of glycerol: chemical toxicity vs osmotic obiology of rat embryos II: A theoretical model for the development
shock at normal and reduced oxygen concentrations. Cryobiology 37, of interrupted slow freezing procedures. Biol. Reprod. 63, 1303–1312.
325–338. doi:10.1006/CRYO.1998.2128 doi:10.1095/BIOLREPROD63.5.1303
Katska-Ksiazkiewicz, L., Lechniak-Cieslak, D., Korwin-Kossakowska, A., Liu, J. L., Kusakabe, H., Chang, C. C., Suzuki, H., and Schmidt, D. W., et al.
Alm, H., Rynska, B., Warzych, E., Sosnowski, J., and Sender, G. (2004). Freeze-dried sperm fertilization leads to full-term development
(2006). Genetical and biotechnological methods of utilization of in rabbits. Biol. Reprod. 70, 1776–1781. doi:10.1095/BIOLREPROD.103.
female reproductive potential in mammals. Reprod. Biol. 6(Suppl. 1), 025957
21–36. Luvoni, G. C. (2000). Current progress on assisted reproduction in dogs
Kawase, Y., Araya, H., Kamada, N., Jishage, K., and Suzuki, H. (2005). and cats: in vitro embryo production. Reprod. Nutr. Dev. 40, 505–512.
Possibility of long-term preservation of freeze-dried mouse spermatozoa. doi:10.1051/RND:2000114
Biol. Reprod. 72, 568–573. doi:10.1095/BIOLREPROD.104.035279 Luvoni, G. C. (2006). Gamete cryopreservation in the domestic cat. The-
Keskintepe, L., Pacholczyk, G., Machnicka, A., Norris, K., Curuk, M. A., riogenology 66, 101–111. doi:10.1016/J.THERIOGENOLOGY.2006.
Khan, I., and Brackett, B. G. (2002). Bovine blastocyst development 03.012
from oocytes injected with freeze-dried spermatozoa. Biol. Reprod. 67, Mamo, S., Bodo, S., Kobolak, J., Polgar, Z., Tolgyesi, G., and Dinnyes, A.
409–415. doi:10.1095/BIOLREPROD67.2.409 (2006). Gene expression profiles of vitrified in vivo derived 8-cell stage
Kimura, Y., and Yanagimachi, R. (1995). Intracytoplasmic sperm mouse embryos detected by high density oligonucleotide microarrays.
injection in the mouse Biol. Reprod. 52, 709–720. doi:10.1095/ Mol. Reprod. Dev. 73, 1380–1392. doi:10.1002/MRD.20588
BIOLREPROD52.4.709 Martino, A., Songsasen, N., and Leibo, S. P. (1996). Development
Kino, K., Pain, B., Leibo, S. P., Cochran, M., Clark, M. E., and Etches, R. J. into blastocysts of bovine oocytes cryopreserved by ultra-rapid cool-
(1997). Production of chicken chimeras from injection of frozen–thawed ing. Biol. Reprod. 54, 1059–1069. doi:10.1095/BIOLREPROD54.
blastodermal cells. Poult. Sci. 76, 753–760. 5.1059
Kobayashi, T., Takeuchi, T., and Yoshizaki, G. (2007). Generation of viable Mascola, L., and Guinan, M. E. (1986). Screening to reduce transmission of
fish from cryopreserved primordial germ cells. Mol. Reprod. Dev. 74, sexually transmitted diseases in semen used for artificial insemination.
207–213. doi:10.1002/MRD.20577 N. Engl. J. Med. 314, 1354–1359.
Kuleshova, L., Gianaroli, L., Magli, C., Ferraretti, A., and Trounson, A. Massip, A., Van Der Zwalmen, P., and Ectors, F. (1987). Recent progress
(1999). Birth following vitrification of a small number of human oocytes: in cryopreservation of cattle embryos. Theriogenology 27, 69–79.
case report. Hum. Reprod. 14, 3077–3079. doi:10.1093/HUMREP/ doi:10.1016/0093-691X(87)90071-9
14.12.3077 Mazur, P. (1963). Kinetics of water loss from cells at subzero temperatures
Kumar, S., Millar, J. D., and Watson, P. F. (2003). The effect of cooling and the likelihood of intracellular freezing. J. Gen. Physiol. 47, 347–369.
rate on the survival of cryopreserved bull, ram, and boar spermatozoa: doi:10.1085/JGP.47.2.347
a comparison of two controlled-rate cooling machines Cryobiology 46, Mazur, P. (1970). Cryobiology: the freezing of biological systems. Science
246–253. doi:10.1016/S0011-2240(03)00040-3 168, 939–949. doi:10.1126/SCIENCE.168.3934.939
Kusakabe, H., Szczygiel, M. A., Whittingham, D. G., and Yanagimachi, R. Mazur, P. (1984). Freezing of living cells: mechanisms and implications. Am.
(2001). Maintenance of genetic integrity in frozen and freeze-dried J. Physiol. 247, C125–C142.
730 Reproduction, Fertility and Development A. Dinnyes et al.

Mazur, P. (1990). Equilibrium, quasi-equilibrium, and non-equilibrium Pukazhenthi, B., Comizzoli, P., Travis, A. J., and Wildt, D. E. (2006a).
freezing of mammalian embryos. Cell Biophys. 17, 53–91. Applications of emerging technologies to the study and conservation
Mazur, P., and Schneider, U. (1986). Osmotic responses of preimplantation of threatened and endangered species. Reprod. Fertil. Dev. 18, 77–90.
mouse and bovine embryos and their cryobiological implications Cell doi:10.1071/RD05117
Biophys. 8, 259–285. Pukazhenthi, B., Laroe, D., Crosier, A., Bush, L. M., Spindler, R., Pelican,
Mazur, P., Rall, W. F., and Leibo, S. P. (1984). Kinetics of water loss and the K. M., Bush, M., Howard, J. G., and Wildt, D. E. (2006b). Challenges in
likelihood of intracellular freezing in mouse ova: Influence of the method cryopreservation of clouded leopard (Neofelis nebulosa) spermatozoa.
of calculating the temperature dependence of water permeability. Cell Theriogenology 66, 1790–1796. doi:10.1016/J.THERIOGENOLOGY.
Biophys. 6, 197–214. 2006.02.020
Moce, E., and Graham, J. K. (2006). Cholesterol-loaded cyclodextrins added Rall, W. F. (1992). Cryopreservation of oocytes and embryos: methods
to fresh bull ejaculates improve sperm cryosurvival. J. Anim. Sci. 84, and applications. Anim. Reprod. Sci. 28, 237–245. doi:10.1016/0378-
826–833. 4320(92)90110-Y
Monfort, S. L., Asher, G. W., Wildt, D. E., Wood, T. C., Schiewe, M. C., Rall, W. F., and Fahy, G. M. (1985). Ice-free cryopreservation of
Williamson, L. R., Bush, M., and Rall, W. F. (1993). Successful intrauter- mouse embryos at −196◦ C by vitrification. Nature 313, 573–575.
ine insemination of Eld’s deer (Cervus eldi thamin) with frozen-thawed doi:10.1038/313573A0
spermatozoa. J. Reprod. Fertil. 99, 459–465. Rho, G. J., Kawarsky, S., Johnson, W. H., Kochhar, K., and Betteridge, K. J.
Mortimer, D. (2004). Current and future concepts and practices in human (1998). Sperm and oocyte treatments to improve the formation of male
sperm cryobanking. Reprod. Biomed. Online 9, 134–151. and female pronuclei and subsequent development following intracyto-
Muldrew, K., and McGann, L. E. (1990). Mechanisms of intracellular ice plasmic sperm injection into bovine oocytes. Biol. Reprod. 59, 918–924.
formation. Biophys. J. 57, 525–532. doi:10.1095/BIOLREPROD59.4.918
Muldrew, K., and McGann, L. E. (1994). The osmotic rupture hypothesis of Robeck, T. R., and O’Brien, J. K. (2004). Effect of cryopreserva-
intracellular freezing injury. Biophys. J. 66, 532–541. tion methods and recryopreservation storage on bottlenose dolphin
Nauk, V. A. (1991). Structure and function of spermatozoa of farm animals (Tursiops truncatus) spermatozoa. Biol. Reprod. 70, 1340–1348.
during cryopreservation. Stiinca Kishinev Moldavia 17, 199[In Russian]. doi:10.1095/BIOLREPROD.103.025304
Nawroth, F., Isachenko, V., Dessole, S., Rahimi, G., and Farina, M., et al. Rodriguez-Martinez, H., Ekwall, H., and Linde-Forsberg, C. (1993). Fine
(2002). Vitrification of human spermatozoa without cryoprotectants. structure and elemental composition of fresh and frozen dog spermato-
Cryo Letters 23, 93–102. zoa. J. Reprod. Fertil. Suppl. 47, 279–285.
Newton, H. (1998). The cryopreservation of ovarian tissue as a strategy Rojas, C., Palomo, M. J., Albarracin, J. L., and Mogas, T. (2004). Vitrification
for preserving the fertility of cancer patients. Hum. Reprod. Update 4, of immature and in vitro matured pig oocytes: study of distribution of
237–247. doi:10.1093/HUMUPD/4.3.237 chromosomes, microtubules, and actin microfilaments. Cryobiology 49,
Ogonuki, N., Mochida, K., Miki, H., Inoue, K., and Fray, M., et al. 211–220. doi:10.1016/J.CRYOBIOL.2004.07.002
(2006). Spermatozoa and spermatids retrieved from frozen repro- Rutherford, A. J., and Gosden, R. G. (1999). Ovarian tissue cryopreservation:
ductive organs or frozen whole bodies of male mice can produce a practical option? Acta Paediatr. Suppl. 88, 13–18.
normal offspring. Proc. Natl Acad. Sci. USA 103, 13 098–13 103. Salamon, S., and Maxwell, W. M. (2000). Storage of ram semen. Anim.
doi:10.1073/PNAS.0605755103 Reprod. Sci. 62, 77–111. doi:10.1016/S0378-4320(00)00155-X
Oktay, K., Newton, H., Aubard, Y., Salha, O., and Gosden, R. G. (1998). Schmidt, D. W., Nedambale, T. L., Kim, C., Maier, D. B., Yang, X. J.,
Cryopreservation of immature human oocytes and ovarian tissue: and Tian, X. C. (2004). Effect of cytoskeleton stabilizing agents on
an emerging technology? Fertil. Steril. 69, 1–7. doi:10.1016/S0015- bovine matured oocytes following vitrification. Fertil. Steril. 82, S26.
0282(97)00207-0 doi:10.1016/J.FERTNSTERT.2004.07.071
Papis, K., Shimizu, M., and Izaike,Y. (2000). Factors affecting the survivabil- Seidel G. E., Jr. (2006). Modifying oocytes and embryos to improve
ity of bovine oocytes vitrified in droplets. Theriogenology 54, 651–658. their cryopreservation. Theriogenology 65, 228–235. doi:10.1016/
doi:10.1016/S0093-691X(00)00380-0 J.THERIOGENOLOGY.2005.09.025
Park, S. E., Son, W. Y., Lee, S. H., Lee, K. A., Ko, J. J., and Cha, K. Y. (1997). Shaw, J. M., Cox, S. L., Trounson, A. O., and Jenkin, G. (2000). Evaluation
Chromosome and spindle configurations of human oocytes matured of the long-term function of cryopreserved ovarian grafts in the mouse,
in vitro after cryopreservation at the germinal vesicle stage. Fertil. Steril. implications for human applications. Mol. Cell. Endocrinol. 161, 103–
68, 920–926. doi:10.1016/S0015-0282(97)00365-8 110. doi:10.1016/S0303-7207(99)00230-0
Paynter, S. J. (2000). Current status of the cryopreservation of Si, W., Zheng, P., Li, Y., Dinnyes, A., and Ji, W. (2004). Effect of glycerol
human unfertilized oocytes. Hum. Reprod. Update 6, 449–456. and dimethyl sulfoxide on cryopreservation of rhesus monkey (Macaca
doi:10.1093/HUMUPD/6.5.449 mulatta) sperm. Am. J. Primatol. 62, 301–306. doi:10.1002/AJP.
Phelps, M. J., Liu, J., Benson, J. D., Willoughby, C. E., Gilmore, J. A., 20023
and Critser, J. K. (1999). Effects of Percoll separation, cryoprotective Smorag, Z., and Gajda, B. (1994). Cryopreservation of mammalian
agents, and temperature on plasma membrane permeability characteris- ova and embryos by vitrification. Biotechnol. Adv. 12, 449–465.
tics of murine spermatozoa and their relevance to cryopreservation Biol. doi:10.1016/0734-9750(94)90019-1
Reprod. 61, 1031–1041. doi:10.1095/BIOLREPROD61.4.1031 Somfai, T., Dinnyes, A., Sage, D., Marosan, M., Carnwath, J. W., Ozawa, M.,
Pitt, R. E., Chandrasekaran, M., and Parks, J. E. (1992). Performance of a Kikuchi, K., and Niemann, H. (2006). Development to the blastocyst
kinetic model for intracellular ice formation based on the extent of super- stage of parthenogenetically activated in vitro matured porcine oocytes
cooling. Cryobiology 29, 359–373. doi:10.1016/0011-2240(92)90037-3 after solid surface vitrification (SSV). Theriogenology 66, 415–422.
Poleo, G. A., Godke, R. R., and Tiersch, T. R. (2005). Intracytoplasmic sperm doi:10.1016/J.THERIOGENOLOGY.2005.11.023
injection using cryopreserved, fixed, and freeze-dried sperm in eggs of Son, W. Y., Park, S. E., Lee, K. A., Lee, W. S., Ko, J. J., Yoon, T. K., and
Nile tilapia. Mar. Biotechnol. (NY) 7, 104–111. doi:10.1007/S10126- Cha, K. Y. (1996). Effects of 1,2-propanediol and freezing–thawing on
004-0162-5 the in vitro developmental capacity of human immature oocytes. Fertil.
Polge, C., Smith, A. U., and Parkes, A. S. (1949). Revival of spermatozoa Steril. 66, 995–999.
after vitrification and dehydratation at low temperatures. Nature 164, Sullivan, R. (2004). Male fertility markers, myth or reality. Anim. Reprod.
666–670. Sci. 82-83, 341–347. doi:10.1016/J.ANIREPROSCI.2004.05.007
Novel gamete storage Reproduction, Fertility and Development 731

Terada, Y., Luetjens, C. M., Sutovsky, P., and Schatten, G. (2000). Wakayama, T., and Yanagimachi, R. (1998). Development of normal mice
Atypical decondensation of the sperm nucleus, delayed replication from oocytes injected with freeze-dried spermatozoa. Nat. Biotechnol.
of the male genome, and sex chromosome positioning following 16, 639–641. doi:10.1038/NBT0798-639
intracytoplasmic human sperm injection (ICSI) into golden hamster Ward, M. A., Kaneko, T., Kusakabe, H., Biggers, J. D., Whittingham, D. G.,
eggs: does ICSI itself introduce chromosomal anomalies? Fertil. Steril. and Yanagimachi, R. (2003). Long-term preservation of mouse sper-
74, 454–460. doi:10.1016/S0015-0282(00)00671-3 matozoa after freeze-drying and freezing without cryoprotection. Biol.
Tharasanit, T., Colenbrander, B., and Stout, T. (2006). Effect of mat- Reprod. 69, 2100–2108. doi:10.1095/BIOLREPROD.103.020529
uration stage at cryopreservation on post-thaw cytoskeleton quality Watson, P. F. (1990).Artificial insemination and the preservation of semen. In
and fertilizability of equine oocytes. Mol. Reprod. Dev. 73, 627–637. ‘Marshall Physiology of Reproduction’. (Ed. G. Lamming.) pp.747–869.
doi:10.1002/MRD.20432 (Churchill Livingston: Edinburgh.)
Thirumala, S., Ferrer, M. S., Al-Jarrah, A., Eilts, B. E., Paccamonti, D. L., Watson, P. F. (1995). Recent developments and concepts in the cryopreser-
and Devireddy, R. V. (2003). Cryopreservation of canine sperma- vation of spermatozoa and the assessment of their post-thawing function
tozoa: theoretical prediction of optimal cooling rates in the pres- Reprod. Fertil. Dev. 7, 871–891. doi:10.1071/RD9950871
ence and absence of cryoprotective agents. Cryobiology 47, 109–124. Watson, P. F. (2000). The causes of reduced fertility with cryopre-
doi:10.1016/J.CRYOBIOL.2003.08.003 served semen. Anim. Reprod. Sci. 60–61, 481–492. doi:10.1016/S0378-
Tominaga, K., and Hamada,Y. (2001). Gel-loading tip as container for vitrifi- 4320(00)00099-3
cation of in vitro-produced bovine embryos. J. Reprod. Dev. 47, 267–273. Wildt, D. E. (2000). Genome resource banking for wildlife research,
doi:10.1262/JRD.47.267 management, and conservation. ILAR J. 41, 228–234.
Toner, M., Cravalho, E. G., and Karel, M. (1990). Thermodynamics and Wildt, D. E., Ellis, S., and Howard, J. G. (2001). Linkage of reproductive
kinetics of intracellular ice formation during freezing of biological cells. sciences: from ‘quick fix’ to ‘integrated’ conservation. J. Reprod. Fertil.
J. Appl. Physiol. 69, 1582–1593. Suppl. 57, 295–307.
Vajta, G., and Kuwayama, M. (2006). Improving cryopreservation systems. Wood, M. J., Candy, C. J., and Holt, W. V. (2001). Gamete and embryo cry-
Theriogenology 65, 236–244. doi:10.1016/J.THERIOGENOLOGY. opreservation in rodents. In ‘Cryobanking the genetic resource: Wildlife
2005.09.026 conservation for the future?’ (Eds P. F. Watson and W. V. Holt) pp.
Vajta, G., Booth, P. J., Holm, P., Greve, T., and Callesen, H. (1997). Successful 227–265. (Taylor & Francis: London.)
vitrification of early stage bovine in vitro produced embryos with the Woods, E. J., Benson, J. D., Agca, Y., and Critser, J. K. (2004). Fundamental
open pulled straw (OPS) method. Cryo Letters 18, 191–195. cryobiology of reproductive cells and tissues. Cryobiology 48, 146–156.
Vajta, G., Holm, P., Kuwayama, M., Booth, P. J., Jacobsen, H., Greve, T., doi:10.1016/J.CRYOBIOL.2004.03.002
and Callesen, H. (1998). Open pulled straw (OPS) vitrification: a new Wu, C., Rui, R., Dai, J., Zhang, C., Ju, S., Xie, B., Lu, X., and Zheng, X.
way to reduce cryoinjuries of bovine ova and embryos. Mol. Reprod. (2006). Effects of cryopreservation on the developmental competence,
Dev. 51, 53–58. doi:10.1002/(SICI)1098-2795(199809)51:1<53::AID- ultrastructure and cytoskeletal structure of porcine oocytes. Mol. Reprod.
MRD6>3.0.CO;2-V Dev. 73, 1454–1462. doi:10.1002/MRD.20579
Van Blerkom, J., and Davis, P. W. (1994). Cytogenetic, cellular, and Yamada, S., Shimazu, Y., Kawano, Y., Nakazawa, M., Naito, K., and
developmental consequences of cryopreservation of immature and Toyoda, Y. (1993). In vitro maturation and fertilization of preovulatory
mature mouse and human oocytes. Microsc. Res. Tech. 27, 165–193. dog oocytes. J. Reprod. Fertil. Suppl. 47, 227–229.
doi:10.1002/JEMT.1070270209 Yoon, T. K., Kim, T. J., Park, S. E., Hong, S. W., Ko, J. J., Chung, H. M.,
Van den Hurk, R., Abir, R., Telfer, E. E., and Bevers, M. M. and Cha, K. Y. (2003). Live births after vitrification of oocytes in
(2000). Primate and bovine immature oocytes and follicles as a stimulated in vitro fertilization-embryo transfer program: freezing
sources of fertilizable oocytes. Hum. Reprod. Update 6, 457–474. immature oocytes. Fertil. Steril. 79, 1323–1326. doi:10.1016/S0015-
doi:10.1093/HUMUPD/6.5.457 0282(03)00258-9
Vishwanath, R., and Shannon, P. (2000). Storage of bovine semen in liquid
and frozen state. Anim. Reprod. Sci. 62, 23–53. doi:10.1016/S0378-
4320(00)00153-6 Manuscript received 26 February 2007, accepted 8 June 2007

http://www.publish.csiro.au/journals/rfd

Вам также может понравиться