SlideShare ist ein Scribd-Unternehmen logo
1 von 7
Downloaden Sie, um offline zu lesen
http://informahealthcare.com/aan 
ISSN: 1939-6368 (print), 1939-6376 (electronic) 
Syst Biol Reprod Med, Early Online: 1–7 
! 2013 Informa Healthcare USA, Inc. DOI: 10.3109/19396368.2013.869273 
REVIEW AND HYPOTHESIS 
Motility, viability, and calcium in the sperm cells 
Jorge Parodi 
Laboratorio de Fisiologı´a de la Reproduccio´n, Escuela de Medicina Veterinaria, Nu´cleo de Investigacio´n en Produccio´n Alimentaria, Facultad de 
Recursos Naturales, Universidad Cato´lica de Temuco, Temuco, Chile 
Abstract 
Sperm cells are complicated in vitro models. Their viability is limited, and physiology is 
complex. The study of their properties is of great application in the animal production as 
viable and functional gametes are essential. It has been shown that the decrease of sperm 
cell viability parallels an increase of the reactive oxygen species (ROS). Reactive oxygen 
species is secondary to normal metabolic processes of the cell-like flagellar movement. 
There is evidence of strategies that reduce ROS levels by using exogenous or endogenous 
antioxidants with the intention that seminal plasma protects the sperm cells and 
increases viability. Perhaps viability can increase by reducing that flagellar movement 
which is regulated by calcium. The phenomenon has not been fully characterized, but it is 
established that in certain mammalian models, the entrance of calcium via specific channels 
such as CATsper or voltage-dependent channels, signals flagellar movement. Previous 
reports have indicated that a change in the concentration of calcium or if the temperature 
is altered, the function of mammal sperm cells is reduced or blocked and viability 
prolonged. Fish sperm can remain immobile for several weeks but when activated the 
number of mobile and viable sperm is reduced at a faster rate. However, if the cells are not 
mobilized the semen can be preserved for longer periods. As presented in this paper, this 
supports the notion that by modulating calcium channels to reduce motility the viability of 
these cells can increase. 
Abbreviations: ROS: reactive oxygen species; ZP: zona pellucid; AR: acrosome reaction; DF: 
disinhibit factor; TEA: tetraethylammonium chloride; CAVs: calcium voltage channels; CatSper: 
cationic sperm 
Keywords 
Calcium, motility, sperm 
History 
Received 4 July 2013 
Revised 2 October 2013 
Accepted 4 October 2013 
Published online 13 December 2013 
Sperm capacitation 
Fertilization is a unique and amazing process involving 
two morphologically distinct cells, the sperm and the oocyte, 
which are recognized and fused together. This process begins 
when the sperm starts to penetrate the oocyte envelope and 
plasma membrane and ends in the exchange of maternal and 
paternal chromosomes, forming the zygote [Patrat et al. 
2006]. The sperm must undergo functional changes following 
its genesis and subsequent maturation in the epididymis. 
Only sperm that have become capacitated can recognize and 
bind to the zona pellucida (ZP). The interaction between the 
sperm and the ZP initiates a signal transduction process 
resulting in exocytosis of the acrosomal contents during the 
acrosome reaction (AR) [Breitbart 2003; Rossato et al. 2001]. 
However, this is only a general picture of the AR phenom-enon, 
and some reports have suggested that an intact ZP is 
not sufficient to induce acrosomal exocytosis [Baibakov 
et al. 2007]. Furthermore, according to the work of 
Dr. Yanagimachi’s group, some mouse sperm passing through 
the cumulus layers are already undergoing or have completed 
the acrosome reaction [Knobil and Neill 1994]. In shrews, the 
acrosome reaction is induced by cumulus cells, but not by the 
ZP [Bedford et al. 2004]. The available evidence suggests a 
general but not unique mechanism of penetration, and it is 
important to consider particular species adaptations when 
manipulating different samples in vitro. The sperm must 
penetrate physical barriers imposed by the oocyte, including 
cumulus oophorus cells, the plasma membrane, and the ZP, 
for which hydrolytic enzymes such as glycohydrolases and 
proteinases are necessary. During capacitation, the sperm 
undergoes functional biochemical and biophysical modifica-tions, 
including changes in the activity of membrane enzymes 
and motility patterns, enabling it to undergo the AR prior to 
fertilization. These modifications include the removal of 
roadblocks to capacitation factors from the sperm surface and 
increased membrane fluidity, cholesterol efflux, intracellular 
Address correspondence to Jorge Parodi, Laboratorio de Fisiologı´a de la 
Reproduccio´n, Escuela de Medicina Veterinaria, Nu´cleo de Investigacio´n 
en Produccio´n Alimentaria, Facultad de Recursos Naturales, Universidad 
Cato´lica de Temuco, Temuco, Chile. Tel: þ56-45-2205564. E-mail: 
jparodi@uct.cl 
Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 
For personal use only.
2 J. Parodi Syst Biol Reprod Med, Early Online: 1–7 
calcium, cAMP, and protein tyrosine phosphorylation 
[Aitken and McLaughlin 2007]. All of these processes 
are regulated by the entry of calcium into the cells, but 
another key factor is the motility of the sperm. The complete 
process had previously been described as a single step: 
the entry of calcium increases motility and the AR. 
However, recently, the calcium wave concept has been 
incorporated (see Figure 1; [Navarrete et al. 2010]), 
indicating that in sperm cells, the first part of the calcium 
wave generates an increase in motility, while the second 
part induces the AR. Furthermore, this process can be 
manipulated while the cells maintain a healthy state [Darszon 
et al. 2011]. 
Capacitation factors 
The reduction of factors inhibiting capacitation (disinhibit 
factor, DF) due to the seminal flow involves a gradual 
release of these factors, from the sperm surface. Their release 
results in a transient state of sperm DF capacitation. This 
ensures the maximum capacity of fertilization at the appro-priate 
location [Acott and Carr 1984; Zhong et al. 1993]. 
Once the DF binds to the sperm surface, it activates a calcium 
ATPase, thus maintaining a low calcium concentration. 
When the DF is released from the sperm surface, an increase 
in intracellular calcium levels is initiated. In vitro studies in 
which the calcium ATPase was inhibited revealed acceler-ation 
of capacitation [Perry et al. 1997]. 
Plasma membrane and ion channels 
The plasma membrane is a lipoprotein interface that acts as a 
permeability barrier allowing the cell to maintain a different 
composition in the intracellular in comparison to the extra-cellular 
medium. The most abundant components of the 
plasma membrane are phospholipids and proteins, which 
together form the fluid mosaic pattern [Hasdemir 2007]. 
The resting potential is a particular state of the membrane 
potential in which the sum of ion currents through the 
membrane is zero. This is due to the presence of transmem-brane 
electrochemical gradients resulting from selective 
permeability to ions and secondary various structures such 
as transmembrane channels, pumps, and ion exchangers. 
From the resting potential, cell excitation can generate an 
action potential that allows the cell to respond to different 
stimuli. During this process, each ion tends to draw the 
membrane potential towards its own electrochemical equilib-rium 
potential (Nernst equation) [Hille 1992]. Ionic currents 
through channels determine transmembrane bioelectric 
phenomena related to the membrane potential in addition 
to modulating enzyme activity, metabolism, and cellular 
genetics activity. Specifically, in sperm cells, the transmem-brane 
ionic currents and their potential, among other factors, 
regulate the intracellular concentration of calcium and the 
genesis of second messengers. These factors are essential for 
fertilization-associated processes, such as sperm motility, 
capacitation, and the AR. Therefore, the study of ion channels 
is extremely valuable for understanding the electrophysio-logical 
processes and biological responses of both excitable 
cells and isolated cells. In particular, determining the roles of 
these channels in the mammalian sperm membrane is 
essential to understand the processes involved in fertilization. 
The main tool for investigating the characteristics and 
distribution of ion channels in the plasma membrane is the 
patch-clamp technique [Neher and Sakmann 1976; Neher 
et al. 1978], which is a high-resolution method that is 
currently used to determine the electrophysiological and 
pharmacological properties of the cell structure. 
Sperm cell viability and function 
One must be careful during the various procedures in which 
sperm are manipulated as alterations can cause premature 
sperm capacitation [Gomez et al. 1997]. This leads to the 
AR impacting the longevity of the sperm. A decrease in 
fertilization capacity can result from the presence of large 
amounts of ROS following ejaculation. Kirchhoff and asso-ciates 
[1998] and Alvarez and Agarwal [2006] indicated 
that sperm produce and export ROS to the extracellular 
environment, most of which are generated by the mitochon-dria, 
secondary to the flagellar activity of the cells. The loss 
of sperm function, i.e., the fertilization capacity, results from 
the presence of high levels of ROS, either following 
ejaculation or secondarily to high levels of motility. These 
studies have indicated that the sperm produced and exported 
ROS to the extracellular environment are the product of the 
monovalent reduction of molecular oxygen during oxidative 
phosphorylation [Alvarez and Agarwal 2006; Kirchhoff et al. 
1998]. Observations made in the laboratory in models 
of immobile sperm cells (salmon or trout) have suggested 
Figure 1. Calcium wave. The upper panel shows a bovine sperm with a 
calcium probe exposed to a high potassium concentration, while the 
lower panel shows a graphic representation of the fluorescence intensity, 
both in control conditions and when sperm are exposed to potassium. 
The figure indicates that there is a wave from the middle piece to the 
head when the sperm are depolarized. Modified figure from Navarrete 
et al. 2010. 
Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 
For personal use only.
DOI: 10.3109/19396368.2013.869273 Sperm cells viability 3 
the existence of long-term viability, lasting for days or weeks, 
while retaining a high rate of fertilization. The common 
feature of such models is the inactive state of the cells, 
without metabolic changes, i.e., ROS production. In mam-malian 
sperm cells, we have observed only a one-hour period 
of viability and function, and these cells show a high motility 
and metabolism. Temperature is important for the regulation 
of cell function, and the preservation and the quality of sperm. 
For example, fish semen display extreme temperature control 
[Alavi and Cosson 2005], and in porcine sperm, temperature 
conservation increases the time of preservation of a sample 
[Althouse et al. 1998]. Furthermore, in fowl, temperature 
regulates calcium influx [Thomson and Wishart 1991]. 
These lines of evidence suggest the importance of tempera-ture 
control during the in vitro manipulation of sperm cells, 
which is correlated with changes during travel in the 
oviduct or in fresh water, in the case of the aquatic species. 
In particular, there may be temperature gradients within the 
oviduct of animals in estrous [Bahat and Eisenbach 2006; 
Hunter and Nichol 1986]. Values presented in the literature 
suggest that these gradients can be on the order of 1–2 C or 
more between the caudal portion of the isthmus and the 
cranial portion of the ampulla in the hours before ovulation. 
This has been proposed to contribute to reducing sperm 
motility and the sperm storage function of the caudal isthmus 
[Hunter and Nichol 1986]. The magnitude of the temperature 
gradient may change according to the stage of the cycle and, 
especially, according to the time of ovulation [Hunter 2012]. 
Therefore, there is a possible influence of temperature on the 
viscosity and viscoelasticity of female tract fluids and on the 
ZP, as in other cell models, membrane viscosity is affected 
by temperature [Stokke et al. 1985]. This factor must be 
considered, and it might be most significant at the time when 
viable spermatozoa are expected to be found in the oviduct 
[Coy et al. 2009]. Temperature is a key factor in the function 
of sperm cells, and we can control it in vitro. Thus, we 
observed natural changes in the oviduct when the sperm cells 
are swimming towards the oocyte. Moreover, in aquatic 
species, environmental conditions are vital to fecundity. 
Kv currents identified in sperm 
A previous study revealed the presence of different types 
and differentially localized potassium channels [Darszon et al. 
2006; Hagiwara and Kawa 1984]. An example is the delayed 
rectifier Kþ type channel found in rat spermatogenic cells, 
which shows a trend that is independent of extracellular 
calcium and is blocked by tetraethylammonium chloride 
(TEA) [Hagiwara and Kawa 1984]. Based on these charac-teristics, 
we identified an inward rectifier Kþ channel referred 
to as Kir [Munoz-Garay et al. 2001]. This channel is also 
regulated by the intracellular pH, with an acidic intracellular 
pH (6.3) inhibiting the current in spermatogenic cells, while a 
rising intracellular pH (7.4) significantly increases conduct-ance 
in these cells. We further identified a third type of Kþ 
channel, designated mSlo3, which was cloned in rat 
spermatogenic cells and has been expressed in Xenopus 
laevis oocytes for biophysical analyses. Recent studies using 
electrophysiological methods allowed an output current from 
the sperm midpiece that is sensitive to TEA to be detected 
[Marconi et al. 2008], and depolarization regulating calcium 
entry was described. 
Regulation of calcium voltage channels (CAVs) 
during capacitation 
During capacitation, ionic channels are susceptible to being 
activated when a change in the configuration of these 
channels occurs and are mediated by a change in the 
membrane potential. In rat and bovine sperm, the membrane 
potential is between 10 and 50mV [Clapham et al. 2003; 
Darszon et al. 2005]. Low voltage calcium is inactivated at 
these voltages and therefore does not respond to depolarizing 
stimuli. Analysis of the membrane potential of rat sperma-tozoa 
showed that only cells that maintain hyperpolarization 
are able to generate an increased flow of calcium secondary 
to contact with the ZP (likely secondary CAVs) and carry 
out the RA [Arnoult et al. 1999]. Capacitation, resulting in 
hyperpolarization, changes the configuration of the CAV in a 
manner that is open to the agonist-mediated ion flow only at 
a specific stage, thus avoiding early RA. Studies in sperm 
conducted using electrophysiological methods have demon-strated 
the role of calcium channel functional are keys in 
capacitation, which are dependent on the membrane potential 
[Darszon et al. 2005; Wennemuth et al. 2000]. However, the 
complete mechanism underlying this phenomenon and its 
regulation via calcium entry is not completely understood. 
In this context, it was recently suggested that calcium entry 
occurs via depolarization and the regulation of motility, with 
a second entry event occurring due to pH regulation and 
depolarization, and this second calcium influx is mediated by 
the AR [Escoffier et al. 2007]. These findings have led to new 
models in which not only the type of CatSper channel is 
responsible for this phenomenon [Xia et al. 2007] but have 
further allowed the electrophysiological investigation of new 
phenomena, such as depolarization, that are also involved in 
the regulation of these voltage-dependent calcium channels. 
Cationic sperm (CatSper) channels 
Four members of the CatSper channels have been described 
(CatSper1-4) in murine sperm [Quill et al. 2001; Ren et al. 
2001]. These channels consist of 6 transmembrane domains 
(6TM1) that are voltage-dependent and calcium-permeable 
and appear to be found only in sperm cells. CatSper1 and 
2 channels have been reported to be essential for sperm 
hyperactivation and fertility. However, reports concerning 
these channels still mainly result from studies of humans and 
mice [Clapham and Garbers 2005]. 
Functional features of the plasma membrane of the sperm 
tail have been described [Ren et al. 2001]. Other reports have 
localized these proteins to the principal piece of the flagellum 
[Kirichok et al. 2006; Qi et al. 2007]. Additional evidence 
regarding the distribution of CatSper in different species and 
its localization in sperm cells is being obtained through 
ongoing investigations, which is important for designing 
solutions for the manipulation of samples. Studies in which 
the expression of this protein has been manipulated have led 
to the generation of a male sterile phenotype in a normal 
mouse model. While the mating behavior, sperm counts, and 
sperm cell morphology of these mutant mice are 
Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 
For personal use only.
4 J. Parodi Syst Biol Reprod Med, Early Online: 1–7 
indistinguishable from those of wild type mice, the CatSper1 
mutant sperm cells are slow, exhibit a reduced basal rate, and 
have no effect on the bathing or the bending of the tail region. 
The mutant sperm cells cannot fertilize the eggs with an intact 
zone pellucid but can fertilize eggs when the outer layers have 
been enzymatically removed [Ren et al. 2001], suggesting 
changes in some cell functions. Male mice lacking CatSper2 
are also infertile due to a lack of the hyperactivated motility 
required for penetration of the extracellular matrix of the 
egg [Quill et al. 2003]. In a study in humans, subfertile men 
with deficient sperm motility showed significantly reduced 
expression of CatSper1 [Nikpoor et al. 2004]. Little is known 
about CatSper3 and CatSper4, but they appear to be involved 
in supporting cell functions in the sperm [Clapham and 
Garbers 2005]. 
The above leads to two questions: (1) can these channels 
explain all of the phenomena observed in the sperm cells? 
(2) is there sufficient evidence to support the idea that 
CatSper channels explain the entire model of the sperm 
activity? It is accepted that CatSper channels and their various 
isoforms are responsible for cellular functions in sperm. 
Additionally, the relationship between CatSper and proges-terone 
has been described, and the authors indicated the effect 
of progesterone on increasing intracellular calcium levels 
[Blackmore 1993; Turner and Meizel 1995]. While the 
relationship between progesterone and CatSper has been 
described [Lishko et al. 2011; Strunker et al. 2011], the 
mechanism underlying the regulation of CatSper function 
by progesterone is not completely understood, although the 
intracellular PI3K-AKT signaling pathway was recently 
implicated in this process. However, progesterone may be 
associated with other receptors in sperm cells, such as 
GABAa [Shi and Roldan 1995], or in the regulation of another 
channel, such as potassium [Kumar et al. 2000], or voltage-dependent 
calcium channels [Bonaccorsi et al. 2001]. 
Progesterone has been described to play a role in the specific 
functions of sperm cell channels [Sagare-Patil et al. 2013]. 
Additionally, CatSper is modulated by pH [Fraire-Zamora and 
Gonzalez-Martinez 2004] and bicarbonate [Wennemuth et al. 
2003]. Nevertheless, additional events must be coordinated 
for fecundation to occur successfully, including the AR, the 
regulation of membrane stability, calcium signaling, and 
mitochondrial function, among others, beyond Catsper modu-lation. 
However, these events are not described in all models, 
and other electrical phenomena can cooperate in the cellular 
events described in sperm. A complete table of ion channels, 
indicating the presence of voltage-dependent calcium chan-nels 
and CatSper, in humans and mice is available [Darszon 
et al. 2011]. This review indicates that we lack a complete 
understanding of the localization of these channels, and there 
are other mechanisms that may alter intracellular calcium. 
Changes observed in the membrane potential 
of sperm cells 
An increase in the membrane potential, described as 
hyperpolarization, occurs during capacitation in rat, bovine, 
and human spermatozoa [Arnoult et al. 1996; Brewis et al. 
2001; Zeng et al. 1996]. In rat sperm, hyperpolarization is the 
result of increased permeability to Kþ [Zeng et al. 1995], 
leading to a change in the membrane potential. During 
capacitation, there is an increase in the pHi of more than 
0.2 units [Zeng et al. 1996], which is sufficient to induce an 
increase of 0.5 to 3 times in the probability of the opening 
of Kir channels found in other tissues [Gutman et al. 2003]. 
Thus, under physiological conditions, an increase in pHi 
activates Kir channels. It has been suggested that this process 
hyperpolarizes the sperm membrane [Krasznai et al. 2000]. 
Furthermore, Kv-activated intracellular calcium is modulated 
by the increase in the concentration of intracellular calcium 
that occurs during capacitation, thus contributing to hyperpo-larization 
[Jagannathan et al. 2002]. Together these observa-tions 
confirm the role of Kþ currents in the hyperpolarization 
of the sperm membrane and its effect on capacitation and the 
subsequent AR. However, in other cell models, the mechan-ism 
reflects blocking the Kþ channel shaft, depolarization, 
and calcium channel opening [Baker et al. 1973; Wellman 
et al. 2001]. In sperm models, it is accepted that Kir channels 
are able to hyperpolarize the membrane, but these channels 
are controlled by physiological phenomena, leading to changes 
in the membrane potential and correcting this potential, 
allowing positive charges to be relocated to restore balance 
and maintain a physiological membrane potential [Gutman 
et al. 2003]. Kv-type channels are present in sperm [Marconi 
et al. 2008], and their current is modulated by peptides, 
suggesting a means to modulate currents in sperm [Parodi 
et al. 2010]. This model is sensitive to ASD and can be applied 
to generate depolarization in other cell models, leading to an 
increase in intracellular calcium levels and consequent cellular 
changes [Navarrete et al. 2010]. Some evidence suggests that 
this mechanism is part of a complex mechanism of regulation 
that also includes the hyperpolarization and depolarization 
described in sperm [Fraire-Zamora and Gonzalez-Martinez 
2004; Gonzalez-Martinez 2003; Neri-Vidaurri Pdel et al. 
2006], which can generate changes in the membrane potential, 
causing an influx of calcium and alterations in the physiology 
of sperm [Babcock and Pfeiffer 1987; Linares-Hernandez et al. 
1998]. It is not hyperpolarization alone that mediates this 
phenomenon. The control of the membrane potential of sperm 
cells can block calcium entry and the associated secondary 
signaling. Many drugs can block changes in the membrane 
potential; could these drugs be used as potential regulators of 
sperm motility? A high concentration of potassium can induce 
changes in intracellular calcium levels, in the form of a wave 
from the middle piece to the head of the sperm. Figure 1 shows 
the effect of high potassium on intracellular calcium levels in 
bovine sperm (from [Navarrete et al. 2010]). 
Calcium as a second messenger 
The processes that generate second messengers that regulate 
cellular physiology have been studied for several years. 
Calcium is important for the regulation of kinase activity, 
phosphatases, gene activation, and protein translation. It is 
required at high concentrations for short periods of time, 
and cells display various mechanisms for finely regulating its 
intracellular concentration and maintaining a physiological 
calcium gradient [Hurwitz 1996; Stewart 1985]. Thus, various 
signals transiently increase intracellular calcium, which is 
indicative of activation of cellular processes, whereas a 
Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 
For personal use only.
DOI: 10.3109/19396368.2013.869273 Sperm cells viability 5 
sustained increase in the cellular concentration indicates cell 
toxicity [Pounds 1984]. These changes in intracellular 
calcium concentrations may vary depending on cell type 
and the type of stimulation involved. Furthermore, they have 
been correlated with the process of vesicular exocytosis 
based on observations made through different techniques for 
measuring calcium currents and cell capacitance [Trifaro 
et al. 2000], including fluorometric measurements of calcium 
levels and amperometric records [Elhamdani et al. 1994]. 
For example, the normal process of vesicle release is 
highly dependent on calcium entry, which is crucial for the 
propagation of nerve impulses and the establishment of neural 
connections responsible for cognitive brain functions. In 
sperm cells, the intracellular activation of vesicles in the 
AR is similar to what is observed in other types of somatic 
cells and depends on changes in calcium levels. These 
findings suggest that different pathways leading to changes 
in calcium levels play a role in the development of different 
models of cell physiology. 
Animal species of industrial interest 
Understanding the influence of reproduction in food produc-tion 
is important in relation to increasing output and yield 
as well as maintaining and preserving genetic markers to 
improve productivity. Regarding the production of meat for 
consumption, cows, goats, pigs, and fish have been instru-mental 
in the development of this industry. In recent years, 
assisted reproduction has begun to be applied in these species 
by preserving oocytes and sperm for later use in artificial 
insemination. The main reference models studied have been 
mice and humans, and similar techniques have been imple-mented 
in cows. Work aimed at the cryopreservation of sperm 
from salmon and other species was recently initiated, with 
sperm being frozen for transport, storage, and handling. There 
is high national and international demand for animal repro-duction, 
as the meat market is steadily increasing, and the 
requirements for animal protein for human populations are 
also increasing [Food and Agriculture Organization of the 
United Nations, 2003]. The world population in 2030 will 
consume more and better food, with 3050 kilocalories (kcal) 
being available per person, compared to 2360 kcal per person/ 
day in the mid-1960s and the 2800 kcal available currently. 
This change reflects the increase in consumption in many 
developing countries, whose average daily intake will be 
approximately 3000 kcal in 2030. For example, it has been 
reported that the domestic consumption of pork per person 
has increased [Oficina de Estudios y Politicas Agrarias, 
2011], reaching values of 23 kg/capita in recent years. Thus, 
pork has become the second most commonly consumed meat, 
while poultry consumption decreased from 2000 to 2006 and 
has remained even at levels of 18 kg/capita over the last 4 
years. The economic returns from the exploitation of animal 
flesh under current market conditions are based on the 
management of their genes and the use of high-genetic value 
players together with the best production techniques to obtain 
high-quality meat products at competitive cost. Reproduction 
is one of the most important aspects of the animal resource, as 
it allows the continuity of the species to be maintained. 
Additionally, the economic importance of reproductive 
behavior in cattle is well-known. Ingvartsen and Moyes 
[2013] summarized that essential studies examining the 
factors that affect the same traits will increase productivity 
in females. Thus, techniques including the control of insem-ination 
have begun to be viewed as an alternative for 
improving production, and the discussion regarding pheno-typic 
traits of importance to the industry is increasing. 
How do we maintain these gametes, increase cell function, 
and apply these techniques under various industrial condi-tions? 
This is not an easy question to answer, but the cellular 
functions of sperm related to generating such compounds as 
well as protocols and conditions applicable in this industry 
should be determined. 
Mature sperm cells are complex cellular machines that 
through a series of steps and environments reach their target, 
the oocyte, and fulfill the purpose of delivering their genetic 
material via fertilization. In this review, we have highlighted 
flagellar motility and capacitation, which is characterized 
by the AR. In recent years, the function of CatSper channels 
as regulatory elements has shown to be indirectly involved in 
modulating the motility and fertilization capacity of sperm as 
well as calcium entry. A recent study has now demonstrated 
that a CatSper channel is involved in the motility but not in 
the AR [Sagare-Patil et al. 2013]. Flagellar movement 
generates various changes, including the production of ROS, 
and these increases can explain the reduction of cell viability. 
Moreover, some sperm cell models can remain immobile for a 
period of time. These sperm cells show a long period of 
viability and maintain their cellular functions for days. When 
activated, the cells become motile upon external signaling 
(i.e., osmotic changes). Calcium regulation is important for 
the general function of cells. In mammalian sperm cells, a 
recent study has suggested that there are two steps regulated 
by calcium entry: first, the motility of sperm cells, and 
second, the AR. Since motility generates ROS it is 
hypothesized here that regulation by calcium reduces the 
motility and the general metabolic state of the cells, leading to 
a reduction of cell mortality. All of these regulatory 
mechanisms are important for the conservation and manipu-lation 
of sperm cells. Because food production, and especially 
that of animal protein, has increased in recent decades, 
reproductive processes must be understood to provide an 
efficient means of control. It is vital for the development of 
the food industry to study these processes, yet little is known 
about the cells involved and the conditions that must occur. 
Thus, we should study other species as a reference for the 
development and maintenance of sperm as a function of 
process. 
Declaration of interest 
The author reports no conflicts of interest. The author alone 
is responsible for the content and writing of the paper. 
References 
Acott, T.S., and Carr, D.W. (1984) Inhibition of bovine spermatozoa by 
caudal epididymal fluid: II. Interaction of pH and a quiescence factor. 
Biol Reprod 30:926–35. 
Aitken, R.J., and McLaughlin, E.A. (2007) Molecular mechanisms 
of sperm capacitation: progesterone-induced secondary calcium 
Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 
For personal use only.
6 J. Parodi Syst Biol Reprod Med, Early Online: 1–7 
oscillations reflect the attainment of a capacitated state. Soc Reprod 
Fertil Suppl 63:273–93. 
Alavi, S.M., and Cosson, J. (2005) Sperm motility in fishes. I. Effects 
of temperature and pH: a review. Cell Biol Int 29:101–10. 
Althouse, G.C., Wilson, M.E., Kuster, C., and Parsley, M. (1998) 
Characterization of lower temperature storage limitations of fresh-extended 
porcine semen. Theriogenology 50:535–43. 
Alvarez, J.G., and Agarwal, A. (2006) Development of a novel home 
sperm test - what are the limitations? Hum Reprod 21:3029–30; 
author reply 3030–3031. 
Arnoult, C., Kazam, I.G., Visconti, P.E., Kopf, G.S., Villaz, M., 
and Florman, H.M. (1999) Control of the low voltage-activated 
calcium channel of mouse sperm by egg ZP3 and by membrane 
hyperpolarization during capacitation. Proc Natl Acad Sci USA 96: 
6757–62. 
Arnoult, C., Zeng, Y., and Florman, H.M. (1996) ZP3-dependent 
activation of sperm cation channels regulates acrosomal secretion 
during mammalian fertilization. J Cell Biol 134:637–45. 
Babcock, D.F., and Pfeiffer, D.R. (1987) Independent elevation of 
cytosolic [Ca2þ] and pH of mammalian sperm by voltage-dependent 
and pH-sensitive mechanisms. J Biol Chem 262:15041–7. 
Bahat, A., and Eisenbach, M. (2006) Sperm thermotaxis. Mol Cell 
Endocrinol 252:115–19. 
Baibakov, B., Gauthier, L., Talbot, P., Rankin, T.L., and Dean, J. (2007) 
Sperm binding to the zona pellucida is not sufficient to induce 
acrosome exocytosis. Development 134:933–43. 
Baker, P.F., Meves, H., and Ridgway, E.B. (1973) Calcium entry in 
response to maintained depolarization of squid axons. J Physiol 231: 
527–48. 
Bedford, J.M., Mock, O.B., and Goodman, S.M. (2004) Novelties of 
conception in insectivorous mammals (Lipotyphla), particularly 
shrews. Biol Rev Camb Philos Soc 79:891–909. 
Blackmore, P.F. (1993) Rapid non-genomic actions of progesterone 
stimulate Ca2þ influx and the acrosome reaction in human sperm. 
Cell Signal 5:531–8. 
Bonaccorsi, L., Forti, G., and Baldi, E. (2001) Low-voltage-activated 
calcium channels are not involved in capacitation and 
biological response to progesterone in human sperm. Int J Androl 24: 
341–51. 
Breitbart, H. (2003) Signaling pathways in sperm capacitation and 
acrosome reaction. Cell Mol Biol (Noisy-le-grand) 49:321–7. 
Brewis, I.A., Morton, I.E., Moore, H.D., and England, G.C. (2001) 
Solubilized zona pellucida proteins and progesterone induce calcium 
influx and the acrosome reaction in capacitated dog spermatozoa. 
Mol Reprod Dev 60:491–7. 
Cabrita, E., Anel, L., and Herraez, M.P. (2001) Effect of external 
cryoprotectants as membrane stabilizers on cryopreserved rainbow 
trout sperm. Theriogenology 56:623–35. 
Clapham, D.E., and Garbers, D.L. (2005) International Union of 
Pharmacology. L. Nomenclature and structure-function relationships 
of CatSper and two-pore channels. Pharmacol Rev 57:451–4. 
Clapham, D.E., Montell, C., Schultz, G., and Julius, D. (2003) 
International Union of Pharmacology. XLIII. Compendium of 
voltage-gated ion channels: transient receptor potential channels. 
Pharmacol Rev 55:591–6. 
Coy, P., Gadea, J., Rath, D., and Hunter, R.H. (2009) Differing sperm 
ability to penetrate the oocyte in vivo and in vitro as revealed using 
colloidal preparations. Theriogenology 72:1171–9. 
Darszon, A., Acevedo, J.J., Galindo, B.E., Hernandez-Gonzalez, E.O., 
Nishigaki, T., Trevino, C.L., et al. (2006) Sperm channel diversity 
and functional multiplicity. Reproduction 131:977–88. 
Darszon, A., Nishigaki, T., Beltran, C., and Trevino, C.L. (2011) 
Calcium channels in the development, maturation, and function of 
spermatozoa. Physiol Rev 91:1305–55. 
Darszon, A., Nishigaki, T., Wood, C., Trevino, C.L., Felix, R., and 
Beltran, C. (2005) Calcium channels and Ca2þ fluctuations in sperm 
physiology. Int Rev Cytol 243:79–172. 
Elhamdani, A., Bossu, J.L., and Feltz, A. (1994) Evolution of the Ca2þ 
current during dialysis of isolated bovine chromaffin cells: effect of 
internal calcium. Cell Calcium 16:357–66. 
Escoffier, J., Boisseau, S., Serres, C., Chen, C.C., Kim, D., Stamboulian, 
S., et al. (2007) Expression, localization and functions in acrosome 
reaction and sperm motility of Ca(V)3.1 and Ca(V)3.2 channels in 
sperm cells: an evaluation from Ca(V)3.1 and Ca(V)3.2 deficient 
mice. J Cell Physiol 212:753–63. 
Food and Agriculture Organization of the United Nations. (2003) Protein 
and amino acid requirements in human nutrition: report of a joint 
FAO/WHO/UNU expert consultation. Author; Geneva. 
Fraire-Zamora, J.J., and Gonzalez-Martinez, M.T. (2004) Effect of 
intracellular pH on depolarization-evoked calcium influx in human 
sperm. Am J Physiol Cell Physiol 287:C1688–96. 
Gomez, M.C., Catt, J.W., Gillan, L., Evans, G., and Maxwell, W.M. 
(1997) Effect of culture, incubation and acrosome reaction of fresh 
and frozen-thawed ram spermatozoa for in vitro fertilization and 
intracytoplasmic sperm injection. Reprod Fertil Dev 9:665–73. 
Gonzalez-Martinez, M.T. (2003) Induction of a sodium-dependent 
depolarization by external calcium removal in human sperm. J Biol 
Chem 278:36304–10. 
Gutman, G.A., Chandy, K.G., Adelman, J.P., Aiyar, J., Bayliss, D.A., 
Clapham, D.E., et al. (2003) International Union of Pharmacology. 
XLI. Compendium of voltage-gated ion channels: potassium channels. 
Pharmacol Rev 55:583–6. 
Hagiwara, S., and Kawa, K. (1984) Calcium and potassium currents 
in spermatogenic cells dissociated from rat seminiferous tubules. 
J Physiol 356:135–49. 
Hasdemir, U. (2007) The role of cell wall organization and active efflux 
pump systems in multidrug resistance of bacteria. Mikrobiyol Bul 41: 
309–27. 
Hille, B. (1992) Ionic channels of excitable membranes. Sinauer; 
Sunderland, MA, Chapter 3, 68p. 
Hunter, R.H. (2012) Temperature gradients in female reproductive 
tissues. Reprod Biomed Online 24:377–80. 
Hunter, R.H., and Nichol, R. (1986) A preovulatory temperature gradient 
between the isthmus and ampulla of pig oviducts during the phase 
of sperm storage. J Reprod Fertil 77:599–606. 
Hurwitz, S. (1996) Homeostatic control of plasma calcium concentra-tion. 
Crit Rev Biochem Mol Biol 31:41–100. 
Ingvartsen, K.L. and Moyes, K. (2013) Nutrition, immune function and 
health of dairy cattle. Animal 7(Suppl 1):112–122. 
Jagannathan, S., Publicover, S.J., and Barratt, C.L. (2002) Voltage-operated 
calcium channels in male germ cells. Reproduction 123: 
203–15. 
Kirchhoff, C., Osterhoff, C., Pera, I., and Schroter, S. (1998) Function of 
human epididymal proteins in sperm maturation. Andrologia 30: 
225–32. 
Kirichok, Y., Navarro, B., and Clapham, D.E. (2006) Whole-cell patch-clamp 
measurements of spermatozoa reveal an alkaline-activated 
Ca2þ channel. Nature 439:737–40. 
Knobil, E. and Neill, J.D. (1994) The Physiology of reproduction. Raven 
Press; New York, 2nd ed, Chapter 1. 
Krasznai, Z., Marian, T., Izumi, H., Damjanovich, S., Balkay, L., 
Tron, L., et al. (2000) Membrane hyperpolarization removes inacti-vation 
of Ca2þ channels, leading to Ca2þ influx and subsequent 
initiation of sperm motility in the common carp. Proc Natl Acad Sci 
USA 97:2052–7. 
Kumar, S., Ying, Y.K., Hong, P., and Maddaiah, V.T. (2000) Potassium 
increases intracellular calcium simulating progesterone action in 
human sperm. Arch Androl 44:93–101. 
Linares-Hernandez, L., Guzman-Grenfell, A.M., Hicks-Gomez, J.J., and 
Gonzalez-Martinez, M.T. (1998) Voltage-dependent calcium influx 
in human sperm assessed by simultaneous optical detection of 
intracellular calcium and membrane potential. Biochim Biophys 
Acta 1372:1–12. 
Lishko, P.V., Botchkina, I.L., and Kirichok, Y. (2011) Progesterone acti-vates 
the principal Ca2þ channel of human sperm. Nature 471:387–91. 
Marconi, M., Sanchez, R., Ulrich, H., and Romero, F. (2008) Potassium 
current in mature bovine spermatozoa. Syst Biol Reprod Med 54: 
231–9. 
Martinez-Lopez, P., Santi, C.M., Trevino, C.L., Ocampo-Gutierrez, A.Y., 
Acevedo, J.J., Alisio, A., et al. (2009) Mouse sperm K+ currents 
stimulated by pH and cAMP possibly coded by Slo3 channels. 
Biochem Biophys Res Commun 381:204–209. 
Munoz-Garay, C., De la Vega-Beltran, J.L., Delgado, R., Labarca, P., 
Felix, R., and Darszon, A. (2001) Inwardly rectifying K(þ) channels 
in spermatogenic cells: functional expression and implication in sperm 
capacitation. Dev Biol 234:261–74. 
Navarrete, P., Martinez-Torres, A., Gutierrez, R.S., Mejia, F.R., and 
Parodi, J. (2010) Venom of the Chilean Latrodectus mactans 
alters bovine spermatozoa calcium and function by blocking the 
TEA-sensitive K(þ) current. Syst Biol Reprod Med 56:303–10. 
Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 
For personal use only.
DOI: 10.3109/19396368.2013.869273 Sperm cells viability 7 
Neher, E., and Sakmann, B. (1976) Noise analysis of drug induced 
voltage clamp currents in denervated frog muscle fibres. J Physiol 
258:705–29. 
Neher, E., Sakmann, B., and Steinbach, J.H. (1978) The extracellular 
patch clamp: a method for resolving currents through individual open 
channels in biological membranes. Pflugers Arch 375:219–28. 
Neri-Vidaurri Pdel, C., Torres-Flores, V., and Gonzalez-Martinez, M.T. 
(2006) A remarkable increase in the pHi sensitivity of volt-age- 
dependent calcium channels occurs in human sperm incubated 
in capacitating conditions. Biochem Biophys Res Commun 343: 
105–9. 
Nikpoor, P., Mowla, S.J., Movahedin, M., Ziaee, S.A., and Tiraihi, T. 
(2004) CatSper gene expression in postnatal development of mouse 
testis and in subfertile men with deficient sperm motility. Hum Reprod 
19:124–8. 
Oficina de Estudios y Politicas Agrarias. (2011) Existencia de cerdos 
en criaderos por tipo, segu´n semestre. Available at: http://www.odepa. 
cl/articulos/MostrarDetalle.action;jsessionid=0144A1D0090E0066E0 
DB26825978CE0C?idcla=12idn=4120 [last accessed 6 Dec 2013]. 
Parodi, J., Navarrete, P., Marconi, M., Gutierrez, R.S., Martinez-Torres, 
A., and Mejias, F.R. (2010) Tetraethylammonium-sensitive K(þ) 
current in the bovine spermatozoa and its blocking by the venom of 
the Chilean Latrodectus mactans. Syst Biol Reprod Med 56:37–43. 
Patrat, C., Auer, J., Fauque, P., Leandri, R.L., Jouannet, P., and Serres, C. 
(2006) Zona pellucida from fertilised human oocytes induces a 
voltage-dependent calcium influx and the acrosome reaction in 
spermatozoa, but cannot be penetrated by sperm. BMC Dev Biol 6:59. 
Perry, R.L., Barratt, C.L., Warren, M.A., and Cooke, I.D. (1997) 
Response of human spermatozoa to an internal calcium ATPase 
inhibitor, 2,5-di(tert-butyl) hydroquinone. J Exp Zool 279:284–90. 
Pounds, J.G. (1984) Effect of lead intoxication on calcium homeostasis 
and calcium-mediated cell function: a review. Neurotoxicology 5: 
295–331. 
Qi, H., Moran, M.M., Navarro, B., Chong, J.A., Krapivinsky, G., 
Krapivinsky, L., et al. (2007) All four CatSper ion channel proteins are 
required for male fertility and sperm cell hyperactivated motility. 
Proc Natl Acad Sci USA 104:1219–23. 
Quill, T.A., Ren, D., Clapham, D.E., and Garbers, D.L. (2001) A voltage-gated 
ion channel expressed specifically in spermatozoa. Proc Natl 
Acad Sci USA 98:12527–31. 
Quill, T.A., Sugden, S.A., Rossi, K.L., Doolittle, L.K., Hammer, R.E., 
and Garbers, D.L. (2003) Hyperactivated sperm motility driven by 
CatSper2 is required for fertilization. Proc Natl Acad Sci USA 100: 
14869–74. 
Ren, D., Navarro, B., Perez, G., Jackson, A.C., Hsu, S., Shi, Q., et al. 
(2001) A sperm ion channel required for sperm motility and male 
fertility. Nature 413:603–9. 
Rossato, M., Di Virgilio, F., Rizzuto, R., Galeazzi, C., and Foresta, C. 
(2001) Intracellular calcium store depletion and acrosome reaction in 
human spermatozoa: role of calcium and plasma membrane potential. 
Mol Hum Reprod 7:119–28. 
Sagare-Patil, V., Vernekar, M., Galvankar, M., and Modi, D. (2013) 
Progesterone utilizes the PI3K-AKT pathway in human spermatozoa 
to regulate motility and hyperactivation but not acrosome reaction. 
Mol Cell Endocrinol 374:82–91. 
Shi, Q.X., and Roldan, E.R. (1995) Evidence that a GABAA-like 
receptor is involved in progesterone-induced acrosomal exocytosis in 
mouse spermatozoa. Biol Reprod 52:373–81. 
Stewart, A.F. (1985) Calcium metabolism without anguish. 
Understanding the body’s homeostatic ‘black box’. Postgrad Med 
77:283–91, 294. 
Stokke, B.T., Mikkelsen, A., and Elgsaeter, A. (1985) Human erythro-cyte 
spectrin dimer intrinsic viscosity: temperature dependence and 
implications for the molecular basis of the erythrocyte membrane free 
energy. Biochim Biophys Acta 816:102–10. 
Strunker, T., Goodwin, N., Brenker, C., Kashikar, N.D., Weyand, I., 
Seifert, R., et al. (2011) The CatSper channel mediates progesterone-induced 
Ca2þ influx in human sperm. Nature 471:382–6. 
Thomson, M.F., and Wishart, G.J. (1991) Temperature-mediated 
regulation of calcium flux and motility in fowl spermatozoa. 
J Reprod Fertil 93:385–91. 
Trifaro, J., Rose, S.D., Lejen, T., and Elzagallaai, A. (2000) Two 
pathways control chromaffin cell cortical F-actin dynamics during 
exocytosis. Biochimie 82:339–52. 
Turner, K.O., and Meizel, S. (1995) Progesterone-mediated efflux of 
cytosolic chloride during the human sperm acrosome reaction. 
Biochem Biophys Res Commun 213:774–80. 
Wellman, G.C., Cartin, L., Eckman, D.M., Stevenson, A.S., 
Saundry, C.M., Lederer, W.J., et al. (2001) Membrane depolarization, 
elevated Ca(2þ) entry, and gene expression in cerebral 
arteries of hypertensive rats. Am J Physiol Heart Circ Physiol 281: 
H2559–67. 
Wennemuth, G., Carlson, A.E., Harper, A.J., and Babcock, D.F. (2003) 
Bicarbonate actions on flagellar and Ca2þ -channel responses: initial 
events in sperm activation. Development 130:1317–26. 
Wennemuth, G., Westenbroek, R.E., Xu, T., Hille, B., and Babcock, D.F. 
(2000) CaV2.2 and CaV2.3 (N- and R-type) Ca2þ channels in 
depolarization-evoked entry of Ca2þ into mouse sperm. J Biol Chem 
275:21210–7. 
Xia, J., Reigada, D., Mitchell, C.H., and Ren, D. (2007) CATSPER 
channel-mediated Ca2þ entry into mouse sperm triggers a tail-to- 
head propagation. Biol Reprod 77:551–9. 
Zeng, Y., Clark, E.N., and Florman, H.M. (1995) Sperm mem-brane 
potential: hyperpolarization during capacitation regu-lates 
zona pellucida-dependent acrosomal secretion. Dev Biol 171: 
554–63. 
Yanagimachi, R. (2011) Mammalian sperm acrosome reaction: 
where does it begin before fertilization? Biol Reprod 85:4–5. 
Zeng, Y., Oberdorf, J.A., and Florman, H.M. (1996) pH regulation 
in mouse sperm: identification of Na(þ)-, Cl()-, and HCO3()- 
dependent and arylaminobenzoate-dependent regulatory mechanisms 
and characterization of their roles in sperm capacitation. Dev Biol 
173:510–20. 
Zhong, C.L., Xin, X.H., and Shi, Q.X. (1993) Inhibition of spermine on 
calcium influx during capacitation of guinea pig spermatozoa in vitro. 
Zhongguo Yao Li Xue Bao 14:141–4. 
Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 
For personal use only.

Weitere ähnliche Inhalte

Was ist angesagt?

Cell cycle presentation by Salman Ul Islam.
Cell cycle presentation by Salman Ul Islam.Cell cycle presentation by Salman Ul Islam.
Cell cycle presentation by Salman Ul Islam.
Salman Ul Islam
 
Cell membrane transport (supplement)
Cell membrane transport (supplement)Cell membrane transport (supplement)
Cell membrane transport (supplement)
ladynoid
 

Was ist angesagt? (20)

Electrical properties and function of membrane ,lipid bilayer and membrane pr...
Electrical properties and function of membrane ,lipid bilayer and membrane pr...Electrical properties and function of membrane ,lipid bilayer and membrane pr...
Electrical properties and function of membrane ,lipid bilayer and membrane pr...
 
Cellular housekeeping
Cellular housekeeping   Cellular housekeeping
Cellular housekeeping
 
Cell cycle and its regulation
Cell cycle and its regulationCell cycle and its regulation
Cell cycle and its regulation
 
CELL CYCLE
CELL CYCLECELL CYCLE
CELL CYCLE
 
Cellular unit in health disease
Cellular unit in health diseaseCellular unit in health disease
Cellular unit in health disease
 
1.6 cell division
1.6 cell division1.6 cell division
1.6 cell division
 
Ribosome
RibosomeRibosome
Ribosome
 
Diffrentiation,Cell diffrentiation,Types of differentiation,Mechanism,Factors...
Diffrentiation,Cell diffrentiation,Types of differentiation,Mechanism,Factors...Diffrentiation,Cell diffrentiation,Types of differentiation,Mechanism,Factors...
Diffrentiation,Cell diffrentiation,Types of differentiation,Mechanism,Factors...
 
Cell cycle presentation by Salman Ul Islam.
Cell cycle presentation by Salman Ul Islam.Cell cycle presentation by Salman Ul Islam.
Cell cycle presentation by Salman Ul Islam.
 
Stem cell technology regenarative medicine
Stem cell technology regenarative medicineStem cell technology regenarative medicine
Stem cell technology regenarative medicine
 
Cell membrane transport (supplement)
Cell membrane transport (supplement)Cell membrane transport (supplement)
Cell membrane transport (supplement)
 
Genetics - 01 A closer look at cells
Genetics - 01 A closer look at cellsGenetics - 01 A closer look at cells
Genetics - 01 A closer look at cells
 
Cell Membrane
Cell MembraneCell Membrane
Cell Membrane
 
Cellular differentiation
Cellular differentiationCellular differentiation
Cellular differentiation
 
CELL CYCLE & A CONCEPT OF CELL THERAPY
CELL CYCLE & A CONCEPT OF CELL THERAPYCELL CYCLE & A CONCEPT OF CELL THERAPY
CELL CYCLE & A CONCEPT OF CELL THERAPY
 
Structural-Functional organization of the Cell
Structural-Functional organization of the CellStructural-Functional organization of the Cell
Structural-Functional organization of the Cell
 
Cell cycle check point By KK Sahu Sir
Cell cycle check point  By KK Sahu SirCell cycle check point  By KK Sahu Sir
Cell cycle check point By KK Sahu Sir
 
1.1 introduction to the cell
1.1 introduction to the cell 1.1 introduction to the cell
1.1 introduction to the cell
 
Ultra structure of cell wall
Ultra structure of cell wallUltra structure of cell wall
Ultra structure of cell wall
 
chemical composition of plasma membrane
chemical composition of plasma membranechemical composition of plasma membrane
chemical composition of plasma membrane
 

Andere mochten auch (7)

espermatozoides 2014
 espermatozoides 2014 espermatozoides 2014
espermatozoides 2014
 
Flores et al 2002 l arginina
Flores et al 2002 l argininaFlores et al 2002 l arginina
Flores et al 2002 l arginina
 
Algo nuevo, algo viejo y algo prestado en alzheimer 2014 jparodi
Algo nuevo, algo viejo y algo prestado en alzheimer 2014 jparodiAlgo nuevo, algo viejo y algo prestado en alzheimer 2014 jparodi
Algo nuevo, algo viejo y algo prestado en alzheimer 2014 jparodi
 
Parodi presentacion valdivia espermatozoides 2014
Parodi presentacion valdivia espermatozoides 2014Parodi presentacion valdivia espermatozoides 2014
Parodi presentacion valdivia espermatozoides 2014
 
Alzheimer 2014 Poro amiloide
Alzheimer 2014 Poro amiloideAlzheimer 2014 Poro amiloide
Alzheimer 2014 Poro amiloide
 
Glucose transport
Glucose transportGlucose transport
Glucose transport
 
Sodium glucose co transporter( SGLT2) Inhibitors
Sodium glucose co transporter( SGLT2) Inhibitors Sodium glucose co transporter( SGLT2) Inhibitors
Sodium glucose co transporter( SGLT2) Inhibitors
 

Ähnlich wie Revision parodi 2013

Boar semen cueva et al 2013
Boar semen cueva et al 2013Boar semen cueva et al 2013
Boar semen cueva et al 2013
Jorge Parodi
 
4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study
4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study
4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study
Stephanie Roberts
 
Describe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And EukaryotesDescribe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And Eukaryotes
Paula Smith
 
s41598-020-71015-9.pdf
s41598-020-71015-9.pdfs41598-020-71015-9.pdf
s41598-020-71015-9.pdf
Hadgi1
 
How butterfly effect or deterministic chaos theory in theorical physics expla...
How butterfly effect or deterministic chaos theory in theorical physics expla...How butterfly effect or deterministic chaos theory in theorical physics expla...
How butterfly effect or deterministic chaos theory in theorical physics expla...
banafsheh61
 
2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materials2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materials
rossbiology
 
Why Proteins Are Essential For Cellular Function
Why Proteins Are Essential For Cellular FunctionWhy Proteins Are Essential For Cellular Function
Why Proteins Are Essential For Cellular Function
Beth Salazar
 
Hypotonic Isotonic And Hypertonic Solutions
Hypotonic Isotonic And Hypertonic SolutionsHypotonic Isotonic And Hypertonic Solutions
Hypotonic Isotonic And Hypertonic Solutions
Kimberly Jones
 
The Discovery Of Vesicle Transportation System Essay
The Discovery Of Vesicle Transportation System EssayThe Discovery Of Vesicle Transportation System Essay
The Discovery Of Vesicle Transportation System Essay
Michelle Singh
 
Physiology at a glance 2013 keyrevisionpoints
Physiology at a glance 2013 keyrevisionpointsPhysiology at a glance 2013 keyrevisionpoints
Physiology at a glance 2013 keyrevisionpoints
Elsa von Licy
 

Ähnlich wie Revision parodi 2013 (20)

Boar semen cueva et al 2013
Boar semen cueva et al 2013Boar semen cueva et al 2013
Boar semen cueva et al 2013
 
4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study
4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study
4. 2 Intracellular Binding Partners Of Podocalyxin Lab Study
 
Describe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And EukaryotesDescribe The Relationship Between Organelles And Eukaryotes
Describe The Relationship Between Organelles And Eukaryotes
 
Aitken 2016 causes and consequences of oxidative stress in spermatozoa
Aitken 2016 causes and consequences of oxidative stress in spermatozoaAitken 2016 causes and consequences of oxidative stress in spermatozoa
Aitken 2016 causes and consequences of oxidative stress in spermatozoa
 
Writing assignment 4 molecular cell biology
Writing assignment 4   molecular cell biologyWriting assignment 4   molecular cell biology
Writing assignment 4 molecular cell biology
 
s41598-020-71015-9.pdf
s41598-020-71015-9.pdfs41598-020-71015-9.pdf
s41598-020-71015-9.pdf
 
How butterfly effect or deterministic chaos theory in theorical physics expla...
How butterfly effect or deterministic chaos theory in theorical physics expla...How butterfly effect or deterministic chaos theory in theorical physics expla...
How butterfly effect or deterministic chaos theory in theorical physics expla...
 
Organelles In Animal Cells Essay
Organelles In Animal Cells EssayOrganelles In Animal Cells Essay
Organelles In Animal Cells Essay
 
2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materials2. Absorption & Secretion Of Materials
2. Absorption & Secretion Of Materials
 
IAS Final Report
IAS Final ReportIAS Final Report
IAS Final Report
 
Why Proteins Are Essential For Cellular Function
Why Proteins Are Essential For Cellular FunctionWhy Proteins Are Essential For Cellular Function
Why Proteins Are Essential For Cellular Function
 
Hypotonic Isotonic And Hypertonic Solutions
Hypotonic Isotonic And Hypertonic SolutionsHypotonic Isotonic And Hypertonic Solutions
Hypotonic Isotonic And Hypertonic Solutions
 
endoplasmic reticulum
endoplasmic reticulum endoplasmic reticulum
endoplasmic reticulum
 
Enzymes
EnzymesEnzymes
Enzymes
 
Enzymes
EnzymesEnzymes
Enzymes
 
Cell migration
Cell migrationCell migration
Cell migration
 
The Discovery Of Vesicle Transportation System Essay
The Discovery Of Vesicle Transportation System EssayThe Discovery Of Vesicle Transportation System Essay
The Discovery Of Vesicle Transportation System Essay
 
Endocytosis and cytoskeleton dynamic
Endocytosis and cytoskeleton dynamicEndocytosis and cytoskeleton dynamic
Endocytosis and cytoskeleton dynamic
 
Physiology at a glance 2013 keyrevisionpoints
Physiology at a glance 2013 keyrevisionpointsPhysiology at a glance 2013 keyrevisionpoints
Physiology at a glance 2013 keyrevisionpoints
 
Biochemistry of Cell Membrane.pptx
Biochemistry of Cell Membrane.pptxBiochemistry of Cell Membrane.pptx
Biochemistry of Cell Membrane.pptx
 

Mehr von Jorge Parodi

Resultados didymo presentacion 2013
Resultados didymo presentacion 2013Resultados didymo presentacion 2013
Resultados didymo presentacion 2013
Jorge Parodi
 
Presentacion curso inb 2012
Presentacion curso inb 2012Presentacion curso inb 2012
Presentacion curso inb 2012
Jorge Parodi
 
Jparodi 2012 molecules and cells
Jparodi 2012 molecules and cellsJparodi 2012 molecules and cells
Jparodi 2012 molecules and cells
Jorge Parodi
 
Presentación nucleo uct 2012 ii
Presentación nucleo uct 2012 iiPresentación nucleo uct 2012 ii
Presentación nucleo uct 2012 ii
Jorge Parodi
 
Presentación nucleo uct 2012
Presentación nucleo uct 2012Presentación nucleo uct 2012
Presentación nucleo uct 2012
Jorge Parodi
 
Presentación uct 2012
Presentación uct 2012Presentación uct 2012
Presentación uct 2012
Jorge Parodi
 
Presentacion ufro 2012
Presentacion ufro 2012Presentacion ufro 2012
Presentacion ufro 2012
Jorge Parodi
 
Inestrosa idn 2011
Inestrosa idn 2011Inestrosa idn 2011
Inestrosa idn 2011
Jorge Parodi
 
In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...
In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...
In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...
Jorge Parodi
 
Movimiento ¿estudiantil
Movimiento ¿estudiantilMovimiento ¿estudiantil
Movimiento ¿estudiantil
Jorge Parodi
 
Aguayo et al 2001 adenosine
Aguayo et al 2001 adenosineAguayo et al 2001 adenosine
Aguayo et al 2001 adenosine
Jorge Parodi
 
Parodi et al 2010 poro amiloide
Parodi et al 2010 poro amiloideParodi et al 2010 poro amiloide
Parodi et al 2010 poro amiloide
Jorge Parodi
 
Parodi et al 2008 veneno ovocitos
Parodi et al 2008 veneno ovocitosParodi et al 2008 veneno ovocitos
Parodi et al 2008 veneno ovocitos
Jorge Parodi
 

Mehr von Jorge Parodi (20)

Cahpter book 2020.pdf
Cahpter book 2020.pdfCahpter book 2020.pdf
Cahpter book 2020.pdf
 
Caligus cinetica 2021.pdf
Caligus cinetica 2021.pdfCaligus cinetica 2021.pdf
Caligus cinetica 2021.pdf
 
Tipos de modelos de cultivos celulares ii
Tipos de modelos de cultivos celulares iiTipos de modelos de cultivos celulares ii
Tipos de modelos de cultivos celulares ii
 
Dolores de la industria salmonicultura en chile v
Dolores de la industria salmonicultura en chile vDolores de la industria salmonicultura en chile v
Dolores de la industria salmonicultura en chile v
 
Articulo TRPC6
Articulo TRPC6Articulo TRPC6
Articulo TRPC6
 
Resultados didymo presentacion 2013
Resultados didymo presentacion 2013Resultados didymo presentacion 2013
Resultados didymo presentacion 2013
 
Jofre et al 2013
Jofre et al 2013Jofre et al 2013
Jofre et al 2013
 
Presentacion curso inb 2012
Presentacion curso inb 2012Presentacion curso inb 2012
Presentacion curso inb 2012
 
Jparodi 2012 molecules and cells
Jparodi 2012 molecules and cellsJparodi 2012 molecules and cells
Jparodi 2012 molecules and cells
 
Presentación nucleo uct 2012 ii
Presentación nucleo uct 2012 iiPresentación nucleo uct 2012 ii
Presentación nucleo uct 2012 ii
 
Presentación nucleo uct 2012
Presentación nucleo uct 2012Presentación nucleo uct 2012
Presentación nucleo uct 2012
 
Presentación uct 2012
Presentación uct 2012Presentación uct 2012
Presentación uct 2012
 
Presentacion ufro 2012
Presentacion ufro 2012Presentacion ufro 2012
Presentacion ufro 2012
 
Inestrosa idn 2011
Inestrosa idn 2011Inestrosa idn 2011
Inestrosa idn 2011
 
In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...
In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...
In tech perforated-patch_clamp_in_non_neuronal_cells_the_model_of_mammalian_s...
 
Arte y ciencia
Arte y cienciaArte y ciencia
Arte y ciencia
 
Movimiento ¿estudiantil
Movimiento ¿estudiantilMovimiento ¿estudiantil
Movimiento ¿estudiantil
 
Aguayo et al 2001 adenosine
Aguayo et al 2001 adenosineAguayo et al 2001 adenosine
Aguayo et al 2001 adenosine
 
Parodi et al 2010 poro amiloide
Parodi et al 2010 poro amiloideParodi et al 2010 poro amiloide
Parodi et al 2010 poro amiloide
 
Parodi et al 2008 veneno ovocitos
Parodi et al 2008 veneno ovocitosParodi et al 2008 veneno ovocitos
Parodi et al 2008 veneno ovocitos
 

Kürzlich hochgeladen

Seismic Method Estimate velocity from seismic data.pptx
Seismic Method Estimate velocity from seismic  data.pptxSeismic Method Estimate velocity from seismic  data.pptx
Seismic Method Estimate velocity from seismic data.pptx
AlMamun560346
 
GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...
GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...
GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...
Lokesh Kothari
 
Formation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksFormation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disks
Sérgio Sacani
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Sérgio Sacani
 
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptxSCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
RizalinePalanog2
 
Pests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdfPests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdf
PirithiRaju
 
Disentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTDisentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOST
Sérgio Sacani
 
Pests of cotton_Borer_Pests_Binomics_Dr.UPR.pdf
Pests of cotton_Borer_Pests_Binomics_Dr.UPR.pdfPests of cotton_Borer_Pests_Binomics_Dr.UPR.pdf
Pests of cotton_Borer_Pests_Binomics_Dr.UPR.pdf
PirithiRaju
 
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsHubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
Sérgio Sacani
 

Kürzlich hochgeladen (20)

TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
TEST BANK For Radiologic Science for Technologists, 12th Edition by Stewart C...
 
Botany 4th semester series (krishna).pdf
Botany 4th semester series (krishna).pdfBotany 4th semester series (krishna).pdf
Botany 4th semester series (krishna).pdf
 
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCRStunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
Stunning ➥8448380779▻ Call Girls In Panchshil Enclave Delhi NCR
 
Seismic Method Estimate velocity from seismic data.pptx
Seismic Method Estimate velocity from seismic  data.pptxSeismic Method Estimate velocity from seismic  data.pptx
Seismic Method Estimate velocity from seismic data.pptx
 
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bNightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
 
GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...
GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...
GUIDELINES ON SIMILAR BIOLOGICS Regulatory Requirements for Marketing Authori...
 
Isotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on IoIsotopic evidence of long-lived volcanism on Io
Isotopic evidence of long-lived volcanism on Io
 
Pulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceuticsPulmonary drug delivery system M.pharm -2nd sem P'ceutics
Pulmonary drug delivery system M.pharm -2nd sem P'ceutics
 
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
 
VIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C PVIRUSES structure and classification ppt by Dr.Prince C P
VIRUSES structure and classification ppt by Dr.Prince C P
 
Formation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksFormation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disks
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
 
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptxSCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
 
Creating and Analyzing Definitive Screening Designs
Creating and Analyzing Definitive Screening DesignsCreating and Analyzing Definitive Screening Designs
Creating and Analyzing Definitive Screening Designs
 
Pests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdfPests of cotton_Sucking_Pests_Dr.UPR.pdf
Pests of cotton_Sucking_Pests_Dr.UPR.pdf
 
Disentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOSTDisentangling the origin of chemical differences using GHOST
Disentangling the origin of chemical differences using GHOST
 
Forensic Biology & Its biological significance.pdf
Forensic Biology & Its biological significance.pdfForensic Biology & Its biological significance.pdf
Forensic Biology & Its biological significance.pdf
 
Vip profile Call Girls In Lonavala 9748763073 For Genuine Sex Service At Just...
Vip profile Call Girls In Lonavala 9748763073 For Genuine Sex Service At Just...Vip profile Call Girls In Lonavala 9748763073 For Genuine Sex Service At Just...
Vip profile Call Girls In Lonavala 9748763073 For Genuine Sex Service At Just...
 
Pests of cotton_Borer_Pests_Binomics_Dr.UPR.pdf
Pests of cotton_Borer_Pests_Binomics_Dr.UPR.pdfPests of cotton_Borer_Pests_Binomics_Dr.UPR.pdf
Pests of cotton_Borer_Pests_Binomics_Dr.UPR.pdf
 
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroidsHubble Asteroid Hunter III. Physical properties of newly found asteroids
Hubble Asteroid Hunter III. Physical properties of newly found asteroids
 

Revision parodi 2013

  • 1. http://informahealthcare.com/aan ISSN: 1939-6368 (print), 1939-6376 (electronic) Syst Biol Reprod Med, Early Online: 1–7 ! 2013 Informa Healthcare USA, Inc. DOI: 10.3109/19396368.2013.869273 REVIEW AND HYPOTHESIS Motility, viability, and calcium in the sperm cells Jorge Parodi Laboratorio de Fisiologı´a de la Reproduccio´n, Escuela de Medicina Veterinaria, Nu´cleo de Investigacio´n en Produccio´n Alimentaria, Facultad de Recursos Naturales, Universidad Cato´lica de Temuco, Temuco, Chile Abstract Sperm cells are complicated in vitro models. Their viability is limited, and physiology is complex. The study of their properties is of great application in the animal production as viable and functional gametes are essential. It has been shown that the decrease of sperm cell viability parallels an increase of the reactive oxygen species (ROS). Reactive oxygen species is secondary to normal metabolic processes of the cell-like flagellar movement. There is evidence of strategies that reduce ROS levels by using exogenous or endogenous antioxidants with the intention that seminal plasma protects the sperm cells and increases viability. Perhaps viability can increase by reducing that flagellar movement which is regulated by calcium. The phenomenon has not been fully characterized, but it is established that in certain mammalian models, the entrance of calcium via specific channels such as CATsper or voltage-dependent channels, signals flagellar movement. Previous reports have indicated that a change in the concentration of calcium or if the temperature is altered, the function of mammal sperm cells is reduced or blocked and viability prolonged. Fish sperm can remain immobile for several weeks but when activated the number of mobile and viable sperm is reduced at a faster rate. However, if the cells are not mobilized the semen can be preserved for longer periods. As presented in this paper, this supports the notion that by modulating calcium channels to reduce motility the viability of these cells can increase. Abbreviations: ROS: reactive oxygen species; ZP: zona pellucid; AR: acrosome reaction; DF: disinhibit factor; TEA: tetraethylammonium chloride; CAVs: calcium voltage channels; CatSper: cationic sperm Keywords Calcium, motility, sperm History Received 4 July 2013 Revised 2 October 2013 Accepted 4 October 2013 Published online 13 December 2013 Sperm capacitation Fertilization is a unique and amazing process involving two morphologically distinct cells, the sperm and the oocyte, which are recognized and fused together. This process begins when the sperm starts to penetrate the oocyte envelope and plasma membrane and ends in the exchange of maternal and paternal chromosomes, forming the zygote [Patrat et al. 2006]. The sperm must undergo functional changes following its genesis and subsequent maturation in the epididymis. Only sperm that have become capacitated can recognize and bind to the zona pellucida (ZP). The interaction between the sperm and the ZP initiates a signal transduction process resulting in exocytosis of the acrosomal contents during the acrosome reaction (AR) [Breitbart 2003; Rossato et al. 2001]. However, this is only a general picture of the AR phenom-enon, and some reports have suggested that an intact ZP is not sufficient to induce acrosomal exocytosis [Baibakov et al. 2007]. Furthermore, according to the work of Dr. Yanagimachi’s group, some mouse sperm passing through the cumulus layers are already undergoing or have completed the acrosome reaction [Knobil and Neill 1994]. In shrews, the acrosome reaction is induced by cumulus cells, but not by the ZP [Bedford et al. 2004]. The available evidence suggests a general but not unique mechanism of penetration, and it is important to consider particular species adaptations when manipulating different samples in vitro. The sperm must penetrate physical barriers imposed by the oocyte, including cumulus oophorus cells, the plasma membrane, and the ZP, for which hydrolytic enzymes such as glycohydrolases and proteinases are necessary. During capacitation, the sperm undergoes functional biochemical and biophysical modifica-tions, including changes in the activity of membrane enzymes and motility patterns, enabling it to undergo the AR prior to fertilization. These modifications include the removal of roadblocks to capacitation factors from the sperm surface and increased membrane fluidity, cholesterol efflux, intracellular Address correspondence to Jorge Parodi, Laboratorio de Fisiologı´a de la Reproduccio´n, Escuela de Medicina Veterinaria, Nu´cleo de Investigacio´n en Produccio´n Alimentaria, Facultad de Recursos Naturales, Universidad Cato´lica de Temuco, Temuco, Chile. Tel: þ56-45-2205564. E-mail: jparodi@uct.cl Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 For personal use only.
  • 2. 2 J. Parodi Syst Biol Reprod Med, Early Online: 1–7 calcium, cAMP, and protein tyrosine phosphorylation [Aitken and McLaughlin 2007]. All of these processes are regulated by the entry of calcium into the cells, but another key factor is the motility of the sperm. The complete process had previously been described as a single step: the entry of calcium increases motility and the AR. However, recently, the calcium wave concept has been incorporated (see Figure 1; [Navarrete et al. 2010]), indicating that in sperm cells, the first part of the calcium wave generates an increase in motility, while the second part induces the AR. Furthermore, this process can be manipulated while the cells maintain a healthy state [Darszon et al. 2011]. Capacitation factors The reduction of factors inhibiting capacitation (disinhibit factor, DF) due to the seminal flow involves a gradual release of these factors, from the sperm surface. Their release results in a transient state of sperm DF capacitation. This ensures the maximum capacity of fertilization at the appro-priate location [Acott and Carr 1984; Zhong et al. 1993]. Once the DF binds to the sperm surface, it activates a calcium ATPase, thus maintaining a low calcium concentration. When the DF is released from the sperm surface, an increase in intracellular calcium levels is initiated. In vitro studies in which the calcium ATPase was inhibited revealed acceler-ation of capacitation [Perry et al. 1997]. Plasma membrane and ion channels The plasma membrane is a lipoprotein interface that acts as a permeability barrier allowing the cell to maintain a different composition in the intracellular in comparison to the extra-cellular medium. The most abundant components of the plasma membrane are phospholipids and proteins, which together form the fluid mosaic pattern [Hasdemir 2007]. The resting potential is a particular state of the membrane potential in which the sum of ion currents through the membrane is zero. This is due to the presence of transmem-brane electrochemical gradients resulting from selective permeability to ions and secondary various structures such as transmembrane channels, pumps, and ion exchangers. From the resting potential, cell excitation can generate an action potential that allows the cell to respond to different stimuli. During this process, each ion tends to draw the membrane potential towards its own electrochemical equilib-rium potential (Nernst equation) [Hille 1992]. Ionic currents through channels determine transmembrane bioelectric phenomena related to the membrane potential in addition to modulating enzyme activity, metabolism, and cellular genetics activity. Specifically, in sperm cells, the transmem-brane ionic currents and their potential, among other factors, regulate the intracellular concentration of calcium and the genesis of second messengers. These factors are essential for fertilization-associated processes, such as sperm motility, capacitation, and the AR. Therefore, the study of ion channels is extremely valuable for understanding the electrophysio-logical processes and biological responses of both excitable cells and isolated cells. In particular, determining the roles of these channels in the mammalian sperm membrane is essential to understand the processes involved in fertilization. The main tool for investigating the characteristics and distribution of ion channels in the plasma membrane is the patch-clamp technique [Neher and Sakmann 1976; Neher et al. 1978], which is a high-resolution method that is currently used to determine the electrophysiological and pharmacological properties of the cell structure. Sperm cell viability and function One must be careful during the various procedures in which sperm are manipulated as alterations can cause premature sperm capacitation [Gomez et al. 1997]. This leads to the AR impacting the longevity of the sperm. A decrease in fertilization capacity can result from the presence of large amounts of ROS following ejaculation. Kirchhoff and asso-ciates [1998] and Alvarez and Agarwal [2006] indicated that sperm produce and export ROS to the extracellular environment, most of which are generated by the mitochon-dria, secondary to the flagellar activity of the cells. The loss of sperm function, i.e., the fertilization capacity, results from the presence of high levels of ROS, either following ejaculation or secondarily to high levels of motility. These studies have indicated that the sperm produced and exported ROS to the extracellular environment are the product of the monovalent reduction of molecular oxygen during oxidative phosphorylation [Alvarez and Agarwal 2006; Kirchhoff et al. 1998]. Observations made in the laboratory in models of immobile sperm cells (salmon or trout) have suggested Figure 1. Calcium wave. The upper panel shows a bovine sperm with a calcium probe exposed to a high potassium concentration, while the lower panel shows a graphic representation of the fluorescence intensity, both in control conditions and when sperm are exposed to potassium. The figure indicates that there is a wave from the middle piece to the head when the sperm are depolarized. Modified figure from Navarrete et al. 2010. Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 For personal use only.
  • 3. DOI: 10.3109/19396368.2013.869273 Sperm cells viability 3 the existence of long-term viability, lasting for days or weeks, while retaining a high rate of fertilization. The common feature of such models is the inactive state of the cells, without metabolic changes, i.e., ROS production. In mam-malian sperm cells, we have observed only a one-hour period of viability and function, and these cells show a high motility and metabolism. Temperature is important for the regulation of cell function, and the preservation and the quality of sperm. For example, fish semen display extreme temperature control [Alavi and Cosson 2005], and in porcine sperm, temperature conservation increases the time of preservation of a sample [Althouse et al. 1998]. Furthermore, in fowl, temperature regulates calcium influx [Thomson and Wishart 1991]. These lines of evidence suggest the importance of tempera-ture control during the in vitro manipulation of sperm cells, which is correlated with changes during travel in the oviduct or in fresh water, in the case of the aquatic species. In particular, there may be temperature gradients within the oviduct of animals in estrous [Bahat and Eisenbach 2006; Hunter and Nichol 1986]. Values presented in the literature suggest that these gradients can be on the order of 1–2 C or more between the caudal portion of the isthmus and the cranial portion of the ampulla in the hours before ovulation. This has been proposed to contribute to reducing sperm motility and the sperm storage function of the caudal isthmus [Hunter and Nichol 1986]. The magnitude of the temperature gradient may change according to the stage of the cycle and, especially, according to the time of ovulation [Hunter 2012]. Therefore, there is a possible influence of temperature on the viscosity and viscoelasticity of female tract fluids and on the ZP, as in other cell models, membrane viscosity is affected by temperature [Stokke et al. 1985]. This factor must be considered, and it might be most significant at the time when viable spermatozoa are expected to be found in the oviduct [Coy et al. 2009]. Temperature is a key factor in the function of sperm cells, and we can control it in vitro. Thus, we observed natural changes in the oviduct when the sperm cells are swimming towards the oocyte. Moreover, in aquatic species, environmental conditions are vital to fecundity. Kv currents identified in sperm A previous study revealed the presence of different types and differentially localized potassium channels [Darszon et al. 2006; Hagiwara and Kawa 1984]. An example is the delayed rectifier Kþ type channel found in rat spermatogenic cells, which shows a trend that is independent of extracellular calcium and is blocked by tetraethylammonium chloride (TEA) [Hagiwara and Kawa 1984]. Based on these charac-teristics, we identified an inward rectifier Kþ channel referred to as Kir [Munoz-Garay et al. 2001]. This channel is also regulated by the intracellular pH, with an acidic intracellular pH (6.3) inhibiting the current in spermatogenic cells, while a rising intracellular pH (7.4) significantly increases conduct-ance in these cells. We further identified a third type of Kþ channel, designated mSlo3, which was cloned in rat spermatogenic cells and has been expressed in Xenopus laevis oocytes for biophysical analyses. Recent studies using electrophysiological methods allowed an output current from the sperm midpiece that is sensitive to TEA to be detected [Marconi et al. 2008], and depolarization regulating calcium entry was described. Regulation of calcium voltage channels (CAVs) during capacitation During capacitation, ionic channels are susceptible to being activated when a change in the configuration of these channels occurs and are mediated by a change in the membrane potential. In rat and bovine sperm, the membrane potential is between 10 and 50mV [Clapham et al. 2003; Darszon et al. 2005]. Low voltage calcium is inactivated at these voltages and therefore does not respond to depolarizing stimuli. Analysis of the membrane potential of rat sperma-tozoa showed that only cells that maintain hyperpolarization are able to generate an increased flow of calcium secondary to contact with the ZP (likely secondary CAVs) and carry out the RA [Arnoult et al. 1999]. Capacitation, resulting in hyperpolarization, changes the configuration of the CAV in a manner that is open to the agonist-mediated ion flow only at a specific stage, thus avoiding early RA. Studies in sperm conducted using electrophysiological methods have demon-strated the role of calcium channel functional are keys in capacitation, which are dependent on the membrane potential [Darszon et al. 2005; Wennemuth et al. 2000]. However, the complete mechanism underlying this phenomenon and its regulation via calcium entry is not completely understood. In this context, it was recently suggested that calcium entry occurs via depolarization and the regulation of motility, with a second entry event occurring due to pH regulation and depolarization, and this second calcium influx is mediated by the AR [Escoffier et al. 2007]. These findings have led to new models in which not only the type of CatSper channel is responsible for this phenomenon [Xia et al. 2007] but have further allowed the electrophysiological investigation of new phenomena, such as depolarization, that are also involved in the regulation of these voltage-dependent calcium channels. Cationic sperm (CatSper) channels Four members of the CatSper channels have been described (CatSper1-4) in murine sperm [Quill et al. 2001; Ren et al. 2001]. These channels consist of 6 transmembrane domains (6TM1) that are voltage-dependent and calcium-permeable and appear to be found only in sperm cells. CatSper1 and 2 channels have been reported to be essential for sperm hyperactivation and fertility. However, reports concerning these channels still mainly result from studies of humans and mice [Clapham and Garbers 2005]. Functional features of the plasma membrane of the sperm tail have been described [Ren et al. 2001]. Other reports have localized these proteins to the principal piece of the flagellum [Kirichok et al. 2006; Qi et al. 2007]. Additional evidence regarding the distribution of CatSper in different species and its localization in sperm cells is being obtained through ongoing investigations, which is important for designing solutions for the manipulation of samples. Studies in which the expression of this protein has been manipulated have led to the generation of a male sterile phenotype in a normal mouse model. While the mating behavior, sperm counts, and sperm cell morphology of these mutant mice are Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 For personal use only.
  • 4. 4 J. Parodi Syst Biol Reprod Med, Early Online: 1–7 indistinguishable from those of wild type mice, the CatSper1 mutant sperm cells are slow, exhibit a reduced basal rate, and have no effect on the bathing or the bending of the tail region. The mutant sperm cells cannot fertilize the eggs with an intact zone pellucid but can fertilize eggs when the outer layers have been enzymatically removed [Ren et al. 2001], suggesting changes in some cell functions. Male mice lacking CatSper2 are also infertile due to a lack of the hyperactivated motility required for penetration of the extracellular matrix of the egg [Quill et al. 2003]. In a study in humans, subfertile men with deficient sperm motility showed significantly reduced expression of CatSper1 [Nikpoor et al. 2004]. Little is known about CatSper3 and CatSper4, but they appear to be involved in supporting cell functions in the sperm [Clapham and Garbers 2005]. The above leads to two questions: (1) can these channels explain all of the phenomena observed in the sperm cells? (2) is there sufficient evidence to support the idea that CatSper channels explain the entire model of the sperm activity? It is accepted that CatSper channels and their various isoforms are responsible for cellular functions in sperm. Additionally, the relationship between CatSper and proges-terone has been described, and the authors indicated the effect of progesterone on increasing intracellular calcium levels [Blackmore 1993; Turner and Meizel 1995]. While the relationship between progesterone and CatSper has been described [Lishko et al. 2011; Strunker et al. 2011], the mechanism underlying the regulation of CatSper function by progesterone is not completely understood, although the intracellular PI3K-AKT signaling pathway was recently implicated in this process. However, progesterone may be associated with other receptors in sperm cells, such as GABAa [Shi and Roldan 1995], or in the regulation of another channel, such as potassium [Kumar et al. 2000], or voltage-dependent calcium channels [Bonaccorsi et al. 2001]. Progesterone has been described to play a role in the specific functions of sperm cell channels [Sagare-Patil et al. 2013]. Additionally, CatSper is modulated by pH [Fraire-Zamora and Gonzalez-Martinez 2004] and bicarbonate [Wennemuth et al. 2003]. Nevertheless, additional events must be coordinated for fecundation to occur successfully, including the AR, the regulation of membrane stability, calcium signaling, and mitochondrial function, among others, beyond Catsper modu-lation. However, these events are not described in all models, and other electrical phenomena can cooperate in the cellular events described in sperm. A complete table of ion channels, indicating the presence of voltage-dependent calcium chan-nels and CatSper, in humans and mice is available [Darszon et al. 2011]. This review indicates that we lack a complete understanding of the localization of these channels, and there are other mechanisms that may alter intracellular calcium. Changes observed in the membrane potential of sperm cells An increase in the membrane potential, described as hyperpolarization, occurs during capacitation in rat, bovine, and human spermatozoa [Arnoult et al. 1996; Brewis et al. 2001; Zeng et al. 1996]. In rat sperm, hyperpolarization is the result of increased permeability to Kþ [Zeng et al. 1995], leading to a change in the membrane potential. During capacitation, there is an increase in the pHi of more than 0.2 units [Zeng et al. 1996], which is sufficient to induce an increase of 0.5 to 3 times in the probability of the opening of Kir channels found in other tissues [Gutman et al. 2003]. Thus, under physiological conditions, an increase in pHi activates Kir channels. It has been suggested that this process hyperpolarizes the sperm membrane [Krasznai et al. 2000]. Furthermore, Kv-activated intracellular calcium is modulated by the increase in the concentration of intracellular calcium that occurs during capacitation, thus contributing to hyperpo-larization [Jagannathan et al. 2002]. Together these observa-tions confirm the role of Kþ currents in the hyperpolarization of the sperm membrane and its effect on capacitation and the subsequent AR. However, in other cell models, the mechan-ism reflects blocking the Kþ channel shaft, depolarization, and calcium channel opening [Baker et al. 1973; Wellman et al. 2001]. In sperm models, it is accepted that Kir channels are able to hyperpolarize the membrane, but these channels are controlled by physiological phenomena, leading to changes in the membrane potential and correcting this potential, allowing positive charges to be relocated to restore balance and maintain a physiological membrane potential [Gutman et al. 2003]. Kv-type channels are present in sperm [Marconi et al. 2008], and their current is modulated by peptides, suggesting a means to modulate currents in sperm [Parodi et al. 2010]. This model is sensitive to ASD and can be applied to generate depolarization in other cell models, leading to an increase in intracellular calcium levels and consequent cellular changes [Navarrete et al. 2010]. Some evidence suggests that this mechanism is part of a complex mechanism of regulation that also includes the hyperpolarization and depolarization described in sperm [Fraire-Zamora and Gonzalez-Martinez 2004; Gonzalez-Martinez 2003; Neri-Vidaurri Pdel et al. 2006], which can generate changes in the membrane potential, causing an influx of calcium and alterations in the physiology of sperm [Babcock and Pfeiffer 1987; Linares-Hernandez et al. 1998]. It is not hyperpolarization alone that mediates this phenomenon. The control of the membrane potential of sperm cells can block calcium entry and the associated secondary signaling. Many drugs can block changes in the membrane potential; could these drugs be used as potential regulators of sperm motility? A high concentration of potassium can induce changes in intracellular calcium levels, in the form of a wave from the middle piece to the head of the sperm. Figure 1 shows the effect of high potassium on intracellular calcium levels in bovine sperm (from [Navarrete et al. 2010]). Calcium as a second messenger The processes that generate second messengers that regulate cellular physiology have been studied for several years. Calcium is important for the regulation of kinase activity, phosphatases, gene activation, and protein translation. It is required at high concentrations for short periods of time, and cells display various mechanisms for finely regulating its intracellular concentration and maintaining a physiological calcium gradient [Hurwitz 1996; Stewart 1985]. Thus, various signals transiently increase intracellular calcium, which is indicative of activation of cellular processes, whereas a Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 For personal use only.
  • 5. DOI: 10.3109/19396368.2013.869273 Sperm cells viability 5 sustained increase in the cellular concentration indicates cell toxicity [Pounds 1984]. These changes in intracellular calcium concentrations may vary depending on cell type and the type of stimulation involved. Furthermore, they have been correlated with the process of vesicular exocytosis based on observations made through different techniques for measuring calcium currents and cell capacitance [Trifaro et al. 2000], including fluorometric measurements of calcium levels and amperometric records [Elhamdani et al. 1994]. For example, the normal process of vesicle release is highly dependent on calcium entry, which is crucial for the propagation of nerve impulses and the establishment of neural connections responsible for cognitive brain functions. In sperm cells, the intracellular activation of vesicles in the AR is similar to what is observed in other types of somatic cells and depends on changes in calcium levels. These findings suggest that different pathways leading to changes in calcium levels play a role in the development of different models of cell physiology. Animal species of industrial interest Understanding the influence of reproduction in food produc-tion is important in relation to increasing output and yield as well as maintaining and preserving genetic markers to improve productivity. Regarding the production of meat for consumption, cows, goats, pigs, and fish have been instru-mental in the development of this industry. In recent years, assisted reproduction has begun to be applied in these species by preserving oocytes and sperm for later use in artificial insemination. The main reference models studied have been mice and humans, and similar techniques have been imple-mented in cows. Work aimed at the cryopreservation of sperm from salmon and other species was recently initiated, with sperm being frozen for transport, storage, and handling. There is high national and international demand for animal repro-duction, as the meat market is steadily increasing, and the requirements for animal protein for human populations are also increasing [Food and Agriculture Organization of the United Nations, 2003]. The world population in 2030 will consume more and better food, with 3050 kilocalories (kcal) being available per person, compared to 2360 kcal per person/ day in the mid-1960s and the 2800 kcal available currently. This change reflects the increase in consumption in many developing countries, whose average daily intake will be approximately 3000 kcal in 2030. For example, it has been reported that the domestic consumption of pork per person has increased [Oficina de Estudios y Politicas Agrarias, 2011], reaching values of 23 kg/capita in recent years. Thus, pork has become the second most commonly consumed meat, while poultry consumption decreased from 2000 to 2006 and has remained even at levels of 18 kg/capita over the last 4 years. The economic returns from the exploitation of animal flesh under current market conditions are based on the management of their genes and the use of high-genetic value players together with the best production techniques to obtain high-quality meat products at competitive cost. Reproduction is one of the most important aspects of the animal resource, as it allows the continuity of the species to be maintained. Additionally, the economic importance of reproductive behavior in cattle is well-known. Ingvartsen and Moyes [2013] summarized that essential studies examining the factors that affect the same traits will increase productivity in females. Thus, techniques including the control of insem-ination have begun to be viewed as an alternative for improving production, and the discussion regarding pheno-typic traits of importance to the industry is increasing. How do we maintain these gametes, increase cell function, and apply these techniques under various industrial condi-tions? This is not an easy question to answer, but the cellular functions of sperm related to generating such compounds as well as protocols and conditions applicable in this industry should be determined. Mature sperm cells are complex cellular machines that through a series of steps and environments reach their target, the oocyte, and fulfill the purpose of delivering their genetic material via fertilization. In this review, we have highlighted flagellar motility and capacitation, which is characterized by the AR. In recent years, the function of CatSper channels as regulatory elements has shown to be indirectly involved in modulating the motility and fertilization capacity of sperm as well as calcium entry. A recent study has now demonstrated that a CatSper channel is involved in the motility but not in the AR [Sagare-Patil et al. 2013]. Flagellar movement generates various changes, including the production of ROS, and these increases can explain the reduction of cell viability. Moreover, some sperm cell models can remain immobile for a period of time. These sperm cells show a long period of viability and maintain their cellular functions for days. When activated, the cells become motile upon external signaling (i.e., osmotic changes). Calcium regulation is important for the general function of cells. In mammalian sperm cells, a recent study has suggested that there are two steps regulated by calcium entry: first, the motility of sperm cells, and second, the AR. Since motility generates ROS it is hypothesized here that regulation by calcium reduces the motility and the general metabolic state of the cells, leading to a reduction of cell mortality. All of these regulatory mechanisms are important for the conservation and manipu-lation of sperm cells. Because food production, and especially that of animal protein, has increased in recent decades, reproductive processes must be understood to provide an efficient means of control. It is vital for the development of the food industry to study these processes, yet little is known about the cells involved and the conditions that must occur. Thus, we should study other species as a reference for the development and maintenance of sperm as a function of process. Declaration of interest The author reports no conflicts of interest. The author alone is responsible for the content and writing of the paper. References Acott, T.S., and Carr, D.W. (1984) Inhibition of bovine spermatozoa by caudal epididymal fluid: II. Interaction of pH and a quiescence factor. Biol Reprod 30:926–35. Aitken, R.J., and McLaughlin, E.A. (2007) Molecular mechanisms of sperm capacitation: progesterone-induced secondary calcium Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 For personal use only.
  • 6. 6 J. Parodi Syst Biol Reprod Med, Early Online: 1–7 oscillations reflect the attainment of a capacitated state. Soc Reprod Fertil Suppl 63:273–93. Alavi, S.M., and Cosson, J. (2005) Sperm motility in fishes. I. Effects of temperature and pH: a review. Cell Biol Int 29:101–10. Althouse, G.C., Wilson, M.E., Kuster, C., and Parsley, M. (1998) Characterization of lower temperature storage limitations of fresh-extended porcine semen. Theriogenology 50:535–43. Alvarez, J.G., and Agarwal, A. (2006) Development of a novel home sperm test - what are the limitations? Hum Reprod 21:3029–30; author reply 3030–3031. Arnoult, C., Kazam, I.G., Visconti, P.E., Kopf, G.S., Villaz, M., and Florman, H.M. (1999) Control of the low voltage-activated calcium channel of mouse sperm by egg ZP3 and by membrane hyperpolarization during capacitation. Proc Natl Acad Sci USA 96: 6757–62. Arnoult, C., Zeng, Y., and Florman, H.M. (1996) ZP3-dependent activation of sperm cation channels regulates acrosomal secretion during mammalian fertilization. J Cell Biol 134:637–45. Babcock, D.F., and Pfeiffer, D.R. (1987) Independent elevation of cytosolic [Ca2þ] and pH of mammalian sperm by voltage-dependent and pH-sensitive mechanisms. J Biol Chem 262:15041–7. Bahat, A., and Eisenbach, M. (2006) Sperm thermotaxis. Mol Cell Endocrinol 252:115–19. Baibakov, B., Gauthier, L., Talbot, P., Rankin, T.L., and Dean, J. (2007) Sperm binding to the zona pellucida is not sufficient to induce acrosome exocytosis. Development 134:933–43. Baker, P.F., Meves, H., and Ridgway, E.B. (1973) Calcium entry in response to maintained depolarization of squid axons. J Physiol 231: 527–48. Bedford, J.M., Mock, O.B., and Goodman, S.M. (2004) Novelties of conception in insectivorous mammals (Lipotyphla), particularly shrews. Biol Rev Camb Philos Soc 79:891–909. Blackmore, P.F. (1993) Rapid non-genomic actions of progesterone stimulate Ca2þ influx and the acrosome reaction in human sperm. Cell Signal 5:531–8. Bonaccorsi, L., Forti, G., and Baldi, E. (2001) Low-voltage-activated calcium channels are not involved in capacitation and biological response to progesterone in human sperm. Int J Androl 24: 341–51. Breitbart, H. (2003) Signaling pathways in sperm capacitation and acrosome reaction. Cell Mol Biol (Noisy-le-grand) 49:321–7. Brewis, I.A., Morton, I.E., Moore, H.D., and England, G.C. (2001) Solubilized zona pellucida proteins and progesterone induce calcium influx and the acrosome reaction in capacitated dog spermatozoa. Mol Reprod Dev 60:491–7. Cabrita, E., Anel, L., and Herraez, M.P. (2001) Effect of external cryoprotectants as membrane stabilizers on cryopreserved rainbow trout sperm. Theriogenology 56:623–35. Clapham, D.E., and Garbers, D.L. (2005) International Union of Pharmacology. L. Nomenclature and structure-function relationships of CatSper and two-pore channels. Pharmacol Rev 57:451–4. Clapham, D.E., Montell, C., Schultz, G., and Julius, D. (2003) International Union of Pharmacology. XLIII. Compendium of voltage-gated ion channels: transient receptor potential channels. Pharmacol Rev 55:591–6. Coy, P., Gadea, J., Rath, D., and Hunter, R.H. (2009) Differing sperm ability to penetrate the oocyte in vivo and in vitro as revealed using colloidal preparations. Theriogenology 72:1171–9. Darszon, A., Acevedo, J.J., Galindo, B.E., Hernandez-Gonzalez, E.O., Nishigaki, T., Trevino, C.L., et al. (2006) Sperm channel diversity and functional multiplicity. Reproduction 131:977–88. Darszon, A., Nishigaki, T., Beltran, C., and Trevino, C.L. (2011) Calcium channels in the development, maturation, and function of spermatozoa. Physiol Rev 91:1305–55. Darszon, A., Nishigaki, T., Wood, C., Trevino, C.L., Felix, R., and Beltran, C. (2005) Calcium channels and Ca2þ fluctuations in sperm physiology. Int Rev Cytol 243:79–172. Elhamdani, A., Bossu, J.L., and Feltz, A. (1994) Evolution of the Ca2þ current during dialysis of isolated bovine chromaffin cells: effect of internal calcium. Cell Calcium 16:357–66. Escoffier, J., Boisseau, S., Serres, C., Chen, C.C., Kim, D., Stamboulian, S., et al. (2007) Expression, localization and functions in acrosome reaction and sperm motility of Ca(V)3.1 and Ca(V)3.2 channels in sperm cells: an evaluation from Ca(V)3.1 and Ca(V)3.2 deficient mice. J Cell Physiol 212:753–63. Food and Agriculture Organization of the United Nations. (2003) Protein and amino acid requirements in human nutrition: report of a joint FAO/WHO/UNU expert consultation. Author; Geneva. Fraire-Zamora, J.J., and Gonzalez-Martinez, M.T. (2004) Effect of intracellular pH on depolarization-evoked calcium influx in human sperm. Am J Physiol Cell Physiol 287:C1688–96. Gomez, M.C., Catt, J.W., Gillan, L., Evans, G., and Maxwell, W.M. (1997) Effect of culture, incubation and acrosome reaction of fresh and frozen-thawed ram spermatozoa for in vitro fertilization and intracytoplasmic sperm injection. Reprod Fertil Dev 9:665–73. Gonzalez-Martinez, M.T. (2003) Induction of a sodium-dependent depolarization by external calcium removal in human sperm. J Biol Chem 278:36304–10. Gutman, G.A., Chandy, K.G., Adelman, J.P., Aiyar, J., Bayliss, D.A., Clapham, D.E., et al. (2003) International Union of Pharmacology. XLI. Compendium of voltage-gated ion channels: potassium channels. Pharmacol Rev 55:583–6. Hagiwara, S., and Kawa, K. (1984) Calcium and potassium currents in spermatogenic cells dissociated from rat seminiferous tubules. J Physiol 356:135–49. Hasdemir, U. (2007) The role of cell wall organization and active efflux pump systems in multidrug resistance of bacteria. Mikrobiyol Bul 41: 309–27. Hille, B. (1992) Ionic channels of excitable membranes. Sinauer; Sunderland, MA, Chapter 3, 68p. Hunter, R.H. (2012) Temperature gradients in female reproductive tissues. Reprod Biomed Online 24:377–80. Hunter, R.H., and Nichol, R. (1986) A preovulatory temperature gradient between the isthmus and ampulla of pig oviducts during the phase of sperm storage. J Reprod Fertil 77:599–606. Hurwitz, S. (1996) Homeostatic control of plasma calcium concentra-tion. Crit Rev Biochem Mol Biol 31:41–100. Ingvartsen, K.L. and Moyes, K. (2013) Nutrition, immune function and health of dairy cattle. Animal 7(Suppl 1):112–122. Jagannathan, S., Publicover, S.J., and Barratt, C.L. (2002) Voltage-operated calcium channels in male germ cells. Reproduction 123: 203–15. Kirchhoff, C., Osterhoff, C., Pera, I., and Schroter, S. (1998) Function of human epididymal proteins in sperm maturation. Andrologia 30: 225–32. Kirichok, Y., Navarro, B., and Clapham, D.E. (2006) Whole-cell patch-clamp measurements of spermatozoa reveal an alkaline-activated Ca2þ channel. Nature 439:737–40. Knobil, E. and Neill, J.D. (1994) The Physiology of reproduction. Raven Press; New York, 2nd ed, Chapter 1. Krasznai, Z., Marian, T., Izumi, H., Damjanovich, S., Balkay, L., Tron, L., et al. (2000) Membrane hyperpolarization removes inacti-vation of Ca2þ channels, leading to Ca2þ influx and subsequent initiation of sperm motility in the common carp. Proc Natl Acad Sci USA 97:2052–7. Kumar, S., Ying, Y.K., Hong, P., and Maddaiah, V.T. (2000) Potassium increases intracellular calcium simulating progesterone action in human sperm. Arch Androl 44:93–101. Linares-Hernandez, L., Guzman-Grenfell, A.M., Hicks-Gomez, J.J., and Gonzalez-Martinez, M.T. (1998) Voltage-dependent calcium influx in human sperm assessed by simultaneous optical detection of intracellular calcium and membrane potential. Biochim Biophys Acta 1372:1–12. Lishko, P.V., Botchkina, I.L., and Kirichok, Y. (2011) Progesterone acti-vates the principal Ca2þ channel of human sperm. Nature 471:387–91. Marconi, M., Sanchez, R., Ulrich, H., and Romero, F. (2008) Potassium current in mature bovine spermatozoa. Syst Biol Reprod Med 54: 231–9. Martinez-Lopez, P., Santi, C.M., Trevino, C.L., Ocampo-Gutierrez, A.Y., Acevedo, J.J., Alisio, A., et al. (2009) Mouse sperm K+ currents stimulated by pH and cAMP possibly coded by Slo3 channels. Biochem Biophys Res Commun 381:204–209. Munoz-Garay, C., De la Vega-Beltran, J.L., Delgado, R., Labarca, P., Felix, R., and Darszon, A. (2001) Inwardly rectifying K(þ) channels in spermatogenic cells: functional expression and implication in sperm capacitation. Dev Biol 234:261–74. Navarrete, P., Martinez-Torres, A., Gutierrez, R.S., Mejia, F.R., and Parodi, J. (2010) Venom of the Chilean Latrodectus mactans alters bovine spermatozoa calcium and function by blocking the TEA-sensitive K(þ) current. Syst Biol Reprod Med 56:303–10. Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 For personal use only.
  • 7. DOI: 10.3109/19396368.2013.869273 Sperm cells viability 7 Neher, E., and Sakmann, B. (1976) Noise analysis of drug induced voltage clamp currents in denervated frog muscle fibres. J Physiol 258:705–29. Neher, E., Sakmann, B., and Steinbach, J.H. (1978) The extracellular patch clamp: a method for resolving currents through individual open channels in biological membranes. Pflugers Arch 375:219–28. Neri-Vidaurri Pdel, C., Torres-Flores, V., and Gonzalez-Martinez, M.T. (2006) A remarkable increase in the pHi sensitivity of volt-age- dependent calcium channels occurs in human sperm incubated in capacitating conditions. Biochem Biophys Res Commun 343: 105–9. Nikpoor, P., Mowla, S.J., Movahedin, M., Ziaee, S.A., and Tiraihi, T. (2004) CatSper gene expression in postnatal development of mouse testis and in subfertile men with deficient sperm motility. Hum Reprod 19:124–8. Oficina de Estudios y Politicas Agrarias. (2011) Existencia de cerdos en criaderos por tipo, segu´n semestre. Available at: http://www.odepa. cl/articulos/MostrarDetalle.action;jsessionid=0144A1D0090E0066E0 DB26825978CE0C?idcla=12idn=4120 [last accessed 6 Dec 2013]. Parodi, J., Navarrete, P., Marconi, M., Gutierrez, R.S., Martinez-Torres, A., and Mejias, F.R. (2010) Tetraethylammonium-sensitive K(þ) current in the bovine spermatozoa and its blocking by the venom of the Chilean Latrodectus mactans. Syst Biol Reprod Med 56:37–43. Patrat, C., Auer, J., Fauque, P., Leandri, R.L., Jouannet, P., and Serres, C. (2006) Zona pellucida from fertilised human oocytes induces a voltage-dependent calcium influx and the acrosome reaction in spermatozoa, but cannot be penetrated by sperm. BMC Dev Biol 6:59. Perry, R.L., Barratt, C.L., Warren, M.A., and Cooke, I.D. (1997) Response of human spermatozoa to an internal calcium ATPase inhibitor, 2,5-di(tert-butyl) hydroquinone. J Exp Zool 279:284–90. Pounds, J.G. (1984) Effect of lead intoxication on calcium homeostasis and calcium-mediated cell function: a review. Neurotoxicology 5: 295–331. Qi, H., Moran, M.M., Navarro, B., Chong, J.A., Krapivinsky, G., Krapivinsky, L., et al. (2007) All four CatSper ion channel proteins are required for male fertility and sperm cell hyperactivated motility. Proc Natl Acad Sci USA 104:1219–23. Quill, T.A., Ren, D., Clapham, D.E., and Garbers, D.L. (2001) A voltage-gated ion channel expressed specifically in spermatozoa. Proc Natl Acad Sci USA 98:12527–31. Quill, T.A., Sugden, S.A., Rossi, K.L., Doolittle, L.K., Hammer, R.E., and Garbers, D.L. (2003) Hyperactivated sperm motility driven by CatSper2 is required for fertilization. Proc Natl Acad Sci USA 100: 14869–74. Ren, D., Navarro, B., Perez, G., Jackson, A.C., Hsu, S., Shi, Q., et al. (2001) A sperm ion channel required for sperm motility and male fertility. Nature 413:603–9. Rossato, M., Di Virgilio, F., Rizzuto, R., Galeazzi, C., and Foresta, C. (2001) Intracellular calcium store depletion and acrosome reaction in human spermatozoa: role of calcium and plasma membrane potential. Mol Hum Reprod 7:119–28. Sagare-Patil, V., Vernekar, M., Galvankar, M., and Modi, D. (2013) Progesterone utilizes the PI3K-AKT pathway in human spermatozoa to regulate motility and hyperactivation but not acrosome reaction. Mol Cell Endocrinol 374:82–91. Shi, Q.X., and Roldan, E.R. (1995) Evidence that a GABAA-like receptor is involved in progesterone-induced acrosomal exocytosis in mouse spermatozoa. Biol Reprod 52:373–81. Stewart, A.F. (1985) Calcium metabolism without anguish. Understanding the body’s homeostatic ‘black box’. Postgrad Med 77:283–91, 294. Stokke, B.T., Mikkelsen, A., and Elgsaeter, A. (1985) Human erythro-cyte spectrin dimer intrinsic viscosity: temperature dependence and implications for the molecular basis of the erythrocyte membrane free energy. Biochim Biophys Acta 816:102–10. Strunker, T., Goodwin, N., Brenker, C., Kashikar, N.D., Weyand, I., Seifert, R., et al. (2011) The CatSper channel mediates progesterone-induced Ca2þ influx in human sperm. Nature 471:382–6. Thomson, M.F., and Wishart, G.J. (1991) Temperature-mediated regulation of calcium flux and motility in fowl spermatozoa. J Reprod Fertil 93:385–91. Trifaro, J., Rose, S.D., Lejen, T., and Elzagallaai, A. (2000) Two pathways control chromaffin cell cortical F-actin dynamics during exocytosis. Biochimie 82:339–52. Turner, K.O., and Meizel, S. (1995) Progesterone-mediated efflux of cytosolic chloride during the human sperm acrosome reaction. Biochem Biophys Res Commun 213:774–80. Wellman, G.C., Cartin, L., Eckman, D.M., Stevenson, A.S., Saundry, C.M., Lederer, W.J., et al. (2001) Membrane depolarization, elevated Ca(2þ) entry, and gene expression in cerebral arteries of hypertensive rats. Am J Physiol Heart Circ Physiol 281: H2559–67. Wennemuth, G., Carlson, A.E., Harper, A.J., and Babcock, D.F. (2003) Bicarbonate actions on flagellar and Ca2þ -channel responses: initial events in sperm activation. Development 130:1317–26. Wennemuth, G., Westenbroek, R.E., Xu, T., Hille, B., and Babcock, D.F. (2000) CaV2.2 and CaV2.3 (N- and R-type) Ca2þ channels in depolarization-evoked entry of Ca2þ into mouse sperm. J Biol Chem 275:21210–7. Xia, J., Reigada, D., Mitchell, C.H., and Ren, D. (2007) CATSPER channel-mediated Ca2þ entry into mouse sperm triggers a tail-to- head propagation. Biol Reprod 77:551–9. Zeng, Y., Clark, E.N., and Florman, H.M. (1995) Sperm mem-brane potential: hyperpolarization during capacitation regu-lates zona pellucida-dependent acrosomal secretion. Dev Biol 171: 554–63. Yanagimachi, R. (2011) Mammalian sperm acrosome reaction: where does it begin before fertilization? Biol Reprod 85:4–5. Zeng, Y., Oberdorf, J.A., and Florman, H.M. (1996) pH regulation in mouse sperm: identification of Na(þ)-, Cl()-, and HCO3()- dependent and arylaminobenzoate-dependent regulatory mechanisms and characterization of their roles in sperm capacitation. Dev Biol 173:510–20. Zhong, C.L., Xin, X.H., and Shi, Q.X. (1993) Inhibition of spermine on calcium influx during capacitation of guinea pig spermatozoa in vitro. Zhongguo Yao Li Xue Bao 14:141–4. Syst Biol Reprod Med Downloaded from informahealthcare.com by 201.186.171.150 on 12/13/13 For personal use only.