Next Article in Journal
Phenotypic Alterations in Erythroid Nucleated Cells of Spleen and Bone Marrow in Acute Hypoxia
Next Article in Special Issue
Thirteen Ovary-Enriched Genes Are Individually Not Essential for Female Fertility in Mice
Previous Article in Journal
A Peripheral Blood Signature of Increased Th1 and Myeloid Cells Combined with Serum Inflammatory Mediators Is Associated with Response to Abatacept in Rheumatoid Arthritis Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sperm-Induced Ca2+ Release in Mammalian Eggs: The Roles of PLCζ, InsP3, and ATP

by
Karl Swann
School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
Cells 2023, 12(24), 2809; https://doi.org/10.3390/cells12242809
Submission received: 25 October 2023 / Revised: 29 November 2023 / Accepted: 6 December 2023 / Published: 10 December 2023
(This article belongs to the Special Issue The Cell Biology of Fertilization)

Abstract

:
Mammalian egg activation at fertilization is triggered by a long-lasting series of increases in cytosolic Ca2+ concentration. These Ca2+ oscillations are due to the production of InsP3 within the egg and the subsequent release of Ca2+ from the endoplasmic reticulum into the cytosol. The generation of InsP3 is initiated by the diffusion of sperm-specific phospholipase Czeta1 (PLCζ) into the egg after gamete fusion. PLCζ enables a positive feedback loop of InsP3 production and Ca2+ release which then stimulates further InsP3 production. Most cytosolic Ca2+ increases in eggs at fertilization involve a fast Ca2+ wave; however, due to the limited diffusion of InsP3, this means that InsP3 must be generated from an intracellular source rather than at the plasma membrane. All mammalian eggs studied generated Ca2+ oscillations in response to PLCζ, but the sensitivity of eggs to PLCζ and to some other stimuli varies between species. This is illustrated by the finding that incubation in Sr2+ medium stimulates Ca2+ oscillations in mouse and rat eggs but not eggs from other mammalian species. This difference appears to be due to the sensitivity of the type 1 InsP3 receptor (IP3R1). I suggest that ATP production from mitochondria modulates the sensitivity of the IP3R1 in a manner that could account for the differential sensitivity of eggs to stimuli that generate Ca2+ oscillations.

1. Introduction

In all studied mammalian eggs, the sperm has been shown to trigger a long-lasting series of increases in cytosolic Ca2+, also referred to as cytosolic Ca2+ oscillations [1,2,3,4]. These Ca2+ oscillations are both necessary and sufficient for activating development. An example recording of Ca2+ oscillations in a fertilizing mouse egg is shown in Figure 1A. Each transient increase in cytosolic Ca2+ in mammalian eggs is the result of Ca2+ release that is stimulated by the production of inositol 1,4,5-trisphosphate (InsP3), which opens the InsP3 receptor (IP3R) in the endoplasmic reticulum (ER).
There is substantial evidence that the Ca2+ oscillations at fertilization are initiated by an isoform of phospholipase C (PLC) called PLCzeta (PLCz1 or PLCζ). PLCζ is a sperm-specific protein that has been shown to cause Ca2+ oscillations and egg activation in mouse, human, pig, and cow eggs [5,6,7]. PLCζ is found in cytosolic sperm extracts that can cause Ca2+ oscillations in eggs after microinjection [7]. The presence of PLCζ in the sperm head can also explain why the direct injection of sperm into eggs, so-called intracytoplasmic sperm injection (ICSI), also triggers a series of prolonged Ca2+ oscillations [8,9]. PLCζ is present in sperm at a concentration that can cause Ca2+ oscillations in eggs, and it is localized inside the sperm in the post-acrosomal region, which is where sperm–egg fusion first occurs [7]. It is thought that the PLCζ protein diffuses into the egg in the first few minutes after sperm–egg membrane fusion.
In this review, I will specifically discuss PLCζ- and InsP3-induced Ca2+ release in mammalian eggs and the ways in which features of eggs make their InsP3-induced responses different from somatic cells. Two of these features involve the mechanism of action of PLCζ, and the third involves the way ATP can modulate Ca2+ release. This review will concentrate on the dynamics and mechanism of sperm-induced Ca2+ release. Other reviews are recommended for a discussion of wider aspects of Ca2+ homeostasis in mammalian eggs [10,11] or else the way in which Ca2+ oscillations stimulate meiotic resumption and embryo development [12,13].
Figure 1. Ca2+ oscillations at fertilization in mouse eggs. Ca2+ was monitored via increases in the fluorescence of the Ca2+-sensitive dye Rhod2-dextran (Rh-D) that was microinjected into eggs as described previously [14]. Part (A) shows the spike-like increases in Ca2+ at fertilization, for which there are typically 10–20 Ca2+ spikes occurring over a period of 3–4 h. In trace (B) is shown a typical example of the first Ca2+ transient that lasts several minutes and consists of smaller oscillations on top of a larger increase.
Figure 1. Ca2+ oscillations at fertilization in mouse eggs. Ca2+ was monitored via increases in the fluorescence of the Ca2+-sensitive dye Rhod2-dextran (Rh-D) that was microinjected into eggs as described previously [14]. Part (A) shows the spike-like increases in Ca2+ at fertilization, for which there are typically 10–20 Ca2+ spikes occurring over a period of 3–4 h. In trace (B) is shown a typical example of the first Ca2+ transient that lasts several minutes and consists of smaller oscillations on top of a larger increase.
Cells 12 02809 g001

2. Diffusion of PLCζ and InsP3 Can Explain PIP2 Distribution and Ca2+ Waves

The initial Ca2+ increase at fertilization in mouse and hamster eggs occurs as a wave of Ca2+ release that crosses the egg. This wave starts from the point of sperm–egg fusion and travels to the opposite side of the egg [3,15]. The time taken for the initial Ca2+ wave in these eggs is about 5–10 s [15]. In subsequent Ca2+ transients, the waves become increasingly rapid, such that after about 15 min, the waves cross the egg in ~1 s and the initiation point is near the cortex, but it varies from one transient to the next [15] (Figure 2).
When initial Ca2+ waves occur, PLCζ will be restricted to the area of sperm–egg fusion. Hence, the initial waves propagate from where PLCζ is concentrated into a cytoplasm that is mostly devoid of PLCζ (see Figure 2). PLCζ is a 70 kDa protein, and the diffusion time for proteins of around this molecular weight across the mouse egg is about 10 min [16]. This means that after ~10 min, PLCζ is expected to have diffused throughout the egg cytoplasm. The spread of PLCζ is coincident with later Ca2+ oscillations which have a rapid rising phase (<1 s) and are known to comprise a Ca2+ wave that travels at 50–100 μm per second (Figure 2). This shift in the pattern of waves has been shown at fertilization in both mouse and hamster eggs [3,15]. The very rapid Ca2+ waves that take over after 10–20 min in either type of egg imply the existence of a positive feedback loop of Ca2+ release. The mechanism of this positive feedback loop helps explain the unusual nature of phosphatidylinositol 1,4,5-bisphoshate (PIP2) distribution in mammalian eggs.
We know that PLCζ hydrolyses PIP2 in eggs to generate InsP3, which causes Ca2+ release [7], but the source of PIP2 is less obvious. Like most other cell types, PIP2 is detected in the plasma membrane of mouse eggs. However, unlike other cell types, much of the PIP2 in mouse eggs is localized in small vesicles that are dispersed throughout the cytoplasm [17,18]. The identity of these vesicles is not clear, but they are likely to be Golgi-derived [18]. The reason why PIP2 in eggs is present in such cytoplasmic vesicles has been unclear, but this localization may be necessary because of a fundamental feature of the mechanism of Ca2+ oscillations in eggs.
There are two classes of models for explaining cytosolic Ca2+ oscillations in cells, and the models differ in the nature of the positive feedback loop [19,20]. One set of models relies upon the ability of the IP3R to open in response to InsP3, and this opening is enhanced by increasing concentrations of Ca2+. The Ca2+-enhanced Ca2+ release operates for the initial phase of Ca2+ release, but IP3R then closes when Ca2+ levels rise to approach the micromolar range. This class of models involves the positive feedback of Ca2+ on theIP3R, and they are therefore referred to as IP3R-based models [19,20]. It has been shown that mouse and human eggs predominantly contain the type 1 IP3R (IP3R1) [21,22], which shows a bell-shaped response to Ca2+ release when stimulated by InsP3; hence, Ca2+ can act to enable positive and negative feedback upon Ca2+ release in eggs [23].
The other set of models involve InsP3-induced Ca2+ release, with Ca2+ then stimulating further PLCζ activity, and this leads to more InsP3 and hence more Ca2+ release. These are the so-called regenerative InsP3 production models. PLCζ is known to be stimulated by Ca2+ at concentrations from around 100 nM to 1 μM [24,25]. PLCζ contains four EF hand domains which are essentially for this high sensitivity to Ca2+ [25]. Hence, PLCζ can provide the basis for such a positive feedback loop of InsP3 production in eggs [24,25]. It should be noted that when InsP3 concentrations are high, the inhibitory effect of Ca2+ on the IP3R is effectively switched off; hence, Ca2+ concentrations can continue to increase to above 1 μM [26]. Either of these two models of oscillation can operate in mammalian eggs and could vary depending upon the stimulus. The critical question is which model applies to Ca2+ oscillations at fertilization or in response to PLCζ.
Fortunately, it is possible to distinguish which mechanism is operating during Ca2+ oscillations in a relatively simple and definitive manner. The mechanism of InsP3-dependent Ca2+ oscillations can be established by applying a pulse of InsP3 during oscillations. This can be carried out in live cells via the photo-release of caged InsP3 [18]. With receptor-based models of regenerative Ca2+ release, a pulse of InsP3 always causes an increase in the frequency of Ca2+ oscillations [19]. In contrast, with Ca2+-induced regenerative InsP3 formation models, a pulse of InsP3 leads to an immediate Ca2+ transient, and this causes a resetting of the phase of oscillations [19]. In practice, what one sees with this model is that a pulse of InsP3 causes a single Ca2+ transient, and the next Ca2+ transient is seen after same interval as all other oscillations, starting from this single transient. Hence, there is no increase in frequency (e.g., Figure 2 in [18]). This means that one sees two very different types of behavior depending upon whether oscillations are based on the IP3R alone or whether they are based upon regenerative InsP3 production. The difference is fundamental to way InsP3 works and does not depend upon the details of the model [19]. It has previously been used to distinguish the different types of Ca2+ oscillations in some somatic cells [19].
We have found that when InsP3 is photo-released in eggs that are oscillating in response to PLCζ, a pulse of InsP3 consistently causes an immediate Ca2+ transient and a resetting of the phase of Ca2+ oscillations, with no increase in frequency (see Figure 2 in Sanders et al. [18]). This shows that when PLCζ is distributed throughout the cytoplasm, Ca2+ oscillations occur with an increasing phase that depends upon Ca2+-induced InsP3 production. Interestingly, this is not true for all types of Ca2+ oscillations in eggs. For example, mouse eggs can also be induced to undergo Ca2+ oscillations via incubation in a Sr2+ medium. With these Sr2+-induced-Ca2+ oscillations, which occur without InsP3 generation, an artificially applied photo-release pulse of InsP3 increases the frequency of Ca2+ oscillations [18]. Hence, Sr2+ causes Ca2+ oscillations in mouse eggs by acting upon the IP3R1 alone, which is consistent with data showing that there are direct stimulatory effects of Sr2+ on IP3R1s in cerebella microsomes [27]. There are clearly two mechanisms of Ca2+ oscillations that can exist in mouse eggs, one for PLCζ and another for Sr2+. Significantly, they demonstrate that PLCζ causes Ca2+ oscillations via a positive feedback mechanism based upon Ca2+-induced InsP3 generation. This means there needs to be a regenerative rise in InsP3 that accompanies the rapid Ca2+ waves that cross the egg in ~1 s.
The rapid generation of InsP3 from PIP2 hydrolysis during the rising phase of Ca2+ increase raises an important issue for large spherical cells like mammalian eggs. In most somatic cell types, PIP2 is predominantly, if not entirely, found in the plasma membrane. Such cells are often small or flat, and InsP3 can diffuse from the plasma membrane to fill the cytoplasm within a few seconds. However, in eggs, if all PIP2 hydrolysis did occur at the plasma membrane, then InsP3 diffusion would set a limit on the propagating speed of the Ca2+ wave.
Determining diffusion coefficients inside cells is not trivial, but the most recent studies suggest a diffusion coefficient of ~100 μm2/s [28]. The diffusion range for a molecule is approximately √2 Dt, where ‘D’ is the diffusion coefficient and ‘t’ is the time scale. Given this, we can estimate that the timescale for InsP3 to travel the 35 μm to the center of a mouse egg will be around 6 s. This is too slow to explain the fast Ca2+ waves that cross the egg in <1 s. Hence, plasma membrane PIP2 hydrolysis will be unable to support the Ca2+ wave that is observed [15]. For the rapid Ca2+ wave to occur, the source of InsP3 production must be much closer to the site of Ca2+ release.
We have modeled Ca2+-induced InsP3 production during the rising phase of a Ca2+ spike in mouse eggs and estimate that the source of InsP3 needs to be within a few microns of the IP3Rs [18]. By labeling intact mouse eggs with a fluorescent probe for PIP2, we found that the PIP2 vesicles are dispersed about 2 microns apart from one another (see Figure 5 in reference [18]). The ER is distributed throughout the egg cytoplasm, so it is likely to be approximately the same distance away from the PIP2 vesicles. Hence, the unusual distribution of PIP2 in cytoplasmic vesicles is consistent with, if not essential for explaining, the known dynamics of oscillations and waves in mouse and hamster eggs. The idea that InsP3 is generated from cytoplasmic PIP2 vesicles is also consistent with the finding that there is no detectable PIP2 hydrolysis in the plasma membrane of mouse eggs during fertilization [17].

3. The Dynamics of InsP3 at Fertilization

The model of PLCζ-induced Ca2+ oscillations in eggs predicts that InsP3 should oscillate in phase with Ca2+. Two studies have reported measurements of InsP3 during Ca2+ oscillations in mouse eggs. The initial study detected InsP3 oscillations in PLCζ-injected mouse eggs as predicted, but only with higher concentrations of PLCζ, and there were no InsP3 increases detected during fertilization [29]. This may have been an issue with the sensitivity of the InsP3 indicator used. A more recent study used a more sensitive InsP3 indicator called IRIS-2.3TMR and detected InsP3 increases during fertilization [30]. It was found that there was a small and monotonic increase in InsP3 during the initial Ca2+ transients, but then distinct oscillations in InsP3 were seen after about 20 min into the series of Ca2+ transients [30].
This shift in pattern is consistent with the diffusion time for PLCζ discussed above. In the first 10–20 min, PLCζ will be localized around the site of sperm fusion, and oscillations will be mainly dependent upon InsP3 diffusion, leading to oscillations dependent upon IP3R stimulation. After 10–20 min, PLCζ will have diffused throughout the egg, and the oscillations are then dependent upon Ca2+-induced InsP3 production, with InsP3 oscillating in synchrony with Ca2+ oscillations (Figure 2). Hence, these experiments are consistent with the idea that an IP3R-based model is operative for the initial Ca2+ transients at fertilization but that after 20 min, the mechanism of regenerative InsP3 production from PIP2 vesicles starts to dominate, and this second mechanism accounts for most of the Ca2+ transients that can occur for the 2–4 h after sperm–egg fusion (Figure 2).
The Ca2+ dependency of PLCζ may also be important in setting the frequency of Ca2+ oscillations over the 3–4 h of the fertilization response. This is because the inter-spike level of Ca2+ will affect the amount of InsP3 that is produced. When fertilizing eggs, or when eggs injected with PLCζ are placed into a Ca2+-free medium, the Ca2+ oscillations slow down or stop [31,32]. It has also been shown that the knockout of TRM7 and Cav3.2 Ca2+ channels in eggs lead to a reduction in the frequency of Ca2+ oscillations at fertilization [33]. These data clearly show that Ca2+ influx is required to maintain Ca2+ oscillations. This result is often interpreted as showing that Ca2+ influx is important for maintaining Ca2+ store content and that the store content plays a role in the timing of each Ca2+ transient. It is clear that Ca2+ will decrease in the ER during each Ca2+ transient in the cytosol, but the rapid cessation of Ca2+ oscillations in a Ca2+-free medium may not be due to ER depletion as such. This is because reducing Ca2+ influx into eggs will also reduce the level of cytosolic Ca2+, and this will then decrease the stimulation of Ca2+-dependent InsP3 generation from PLCζ. Hence, in a Ca2+-free medium, there will be both a lower ER Ca2+ concentration and a lower cytosolic Ca2+ concentration.
The relative importance of Ca2+ store content versus cytosolic Ca2+ level can be distinguished via a simple experiment using the ER Ca2+ pump inhibitor thapsigargin [18]. Applying low concentrations of thapsigargin can be used to reduce the Ca2+ store content whilst increasing cytosolic Ca2+ levels. If mouse eggs are induced to undergo Ca2+ oscillations via the injection of PLCζ, the oscillations can be slowed down, or stopped, by placing them in a Ca2+-free medium. When low concentrations of thapsigargin are then added to these eggs (which are still in a Ca2+-free medium), Ca2+ oscillations can be restarted, and the oscillations can continue for more than 1 h [18]. During these oscillations, there is a small increase in cytosolic Ca2+ levels and a slight depletion of Ca2+ stores, as shown by a smaller amplitude of Ca2+ spikes. However, the data are remarkable in that they show that mouse eggs can undergo Ca2+ oscillations for more than an hour in a Ca2+-free medium if cytosolic Ca2+ is elevated. We know that the activity of PLCζ is very sensitive to increases in Ca2+ around the 100–500 nM range [24] and that thapsigargin causes a small increase cytosolic Ca2+ within this range [17]. Hence, the simplest explanation for this data is that the ability of cytosolic Ca2+ to stimulate PLCζ can play a key role in triggering each Ca2+ spike and that the Ca2+ store content may be less significant. This result also implies that it requires more than an hour of oscillations for Ca2+ stores to become depleted of Ca2+ to the point that no more release occurs.

4. Models of Ca2+ Oscillations in Eggs

There are many mathematical models of intracellular Ca2+ oscillations in somatic cells [19,20]. We have a good understanding of how the sperm causes Ca2+ release in mammalian eggs, but there are no specific mathematical models for mammalian eggs, and some features have yet to be explained in the context of existing generic models of Ca2+ oscillations.
One distinctive feature in mouse eggs, shown in Figure 1B, is that the first Ca2+ spike usually has multiple spikes on top of a larger, longer Ca2+ increase. This was seen in some of the first Ca2+ recordings of Ca2+ oscillations, which used aequorin to measure Ca2+, and we see it in most of our recordings using Ca2+ dyes at fertilization and with PLCζ [1,7]. These small oscillations on top of the first spike are a feature of some models, but we do not know how they occur in eggs.
In addition, we find that there is often a transient increase in the frequency of Ca2+ oscillations at about an hour after sperm–egg fusion (Figure 1A). We see this using microinjected dextran-linked Ca2+ dyes to measure Ca2+, but we did not see with earlier recordings using AM-loaded Ca2+ dyes which may be less reliable because they are associated with formaldehyde generation [34]. Most significantly, it is not clear how each Ca2+ increase is terminated. We can explain the regenerative rise in terms of Ca2+ stimulating PLCζ [18], but the nature of the negative feedback that decreases Ca2+ levels may require some rethinking. Some models of Ca2+ oscillations invoke the complete emptying of Ca2+ stores to terminate Ca2+ release, but this does not occur during each Ca2+ transient in mouse eggs [18,32]. Most models invoke a Ca2+-induced desensitization of the IP3R to terminate Ca2+ release [19,20]. However, when InsP3 concentrations are high, which is expected with PLCζ, Ca2+ is not effective in closing the IP3R [26]. Ca2+-induced desensitization of IP3Rs may not occur physiologically for most Ca2+ transients in fertilizing mammalian eggs.

5. PLCζ and Other Sperm Factors

PLCζ has been shown to cause Ca2+ oscillations when injected as RNA or protein into mouse, human, pig, or cow eggs. Such studies have involved many different research groups; hence, there are multiple independent replications of the ability of PLCζ to cause Ca2+ oscillations. However, there is also evidence that there might be another sperm factor in mammalian fertilization. This is because when mouse eggs are fertilized by PLCζ KO sperm, there are still somewhere between one and four Ca2+ spikes that occur about an hour after sperm–egg fusion [9,35]. It is entirely unclear how sperm can trigger these Ca2+ oscillations. It is possible that the sperm could introduce a second factor into the egg that causes InsP3 production and Ca2+ release.
There are some sperm factor candidates that have been proposed as possible alternatives or additional factors to PLCζ. These other sperm factors include a protein called PAWP and an extramitochondrial form of citrate synthase [36,37,38]. Both proteins have been suggested to be capable of causing Ca2+ release and egg activation in mammalian eggs. We carried out a series of experiments microinjecting PAWP RNA or PAWP protein into mouse eggs. We also tested the effects peptides of from PAWP that were reported to block Ca2+ oscillations at fertilization. We could not reproduce any of the original findings on PAWP and found no evidence that it causes Ca2+ release [39,40]. We have also injected citrate synthase (which is commercially available) into mouse eggs and found no sign of Ca2+ release. Consequently, a problem with these other candidate sperm factors is that the primary studies are not readily reproducible. Until independent laboratories can reproduce the claimed effects of PAWP or citrate synthase, it is difficult to assess their role in fertilization. Such failures to reproduce key observations contrast with the numerous independent studies on the effects of PLCζ.
Whatever the nature of a putative second sperm factor, there are some characteristics of Ca2+ release in eggs fertilized by PLCζ KO sperm that need to be explained. For example, when PLCζ KO sperm are injected into mouse eggs (as in ICSI), there are no Ca2+ oscillations [9,35]. However, as noted above, PLCζ KO sperm cause 1–4 Ca2+ spikes after sperm–egg fusion in IVF [9]. Hence, the second factor works in IVF but not in ICSI. It is not known why the putative second factor only operates during normal IVF. Also, in IVF with PLCζ KO sperm, there is always a distinct delay of about 1 h between sperm–egg fusion and the initiation of Ca2+ oscillations. It is again unclear why it should take about one hour for a putative second factor to trigger Ca2+ release. Hopefully, the identification of a ‘second sperm factor’ will provide an explanation for these characteristics.

6. ATP and the Sensitivity of Ca2+ Release in Eggs

One feature of Ca2+ release in mammalian eggs that is rarely discussed is the way that eggs from different species differ in the ability to undergo Ca2+ oscillations. This was evident from early studies on hamster and mouse eggs. For example, inserting a micropipette to cause a sustained injection of InsP3 causes a heavily damped series of Ca2+ oscillations in hamster eggs that typically stop after 10–15 min [41]. However, the same type of experiment with the sustained injection of InsP3 into mouse eggs leads to undamped and prolonged Ca2+ oscillations, even with tenfold lower concentrations of InsP3 [42]. We later found that mouse eggs are much more sensitive to human PLCζ than human eggs [43]. The difference in sensitivity to sperm PLCζ is at least 30-fold, which is more than can be accounted for by the size difference between mouse and human eggs.
The difference in sensitivity between eggs of different species is most evident with Sr2+ which, as noted above, stimulates the IP3R1 to cause Ca2+ release. Incubation in a Sr2+ medium is very effective and reliable in causing prolonged Ca2+ oscillations in mouse eggs and rat eggs [4,18,44,45]. However, under the same conditions, human eggs do not show any Ca2+ release in response to a Sr2+ medium [44,46].
It is not clear why this difference exists because mouse and human eggs have the same IP3R1 and the same plasma membrane TrV3 channels that allow Sr2+ to enter the egg [44,47,48]. Despite the simplicity and significance of the experiment, it is notable that there are no reports for Sr2+-induced Ca2+ oscillations in pig, cow, or hamster eggs. These data suggest that mouse and rat eggs have much more sensitive Ca2+ release than eggs from humans and hamsters and probably compared to eggs from pigs and cattle.
It has not been clear why eggs from different mammalian species differ in their sensitivity to PLCζ, InsP3, and Sr2+, but the underlying difference appears to be due to the IP3R. We have tested the sensitivity of eggs using caged InsP3 and UV light to deliver precise pulses of InsP3. We found that mouse eggs are about 10 times more sensitive to InsP3-induced Ca2+ release than human eggs [46]. The implication is that something in mouse eggs is sensitizing IP3R1s in a way that is not found in human eggs. There may be some subtle difference in the structure of the IP3R1 between mouse and human eggs, but it is not known what this might be. The IP3R can be modulated by cycle cell protein kinases [11], but it is not obvious why this would be different between mouse and human eggs since they are both arrested at the metaphase of second meiosis. We recently presented data that suggest the relevant factor that regulates the IP3Rs in mammalian eggs is the concentration of ATP.
Most somatic cells express combinations of type 2 or 3 IP3Rs, but all mammalian eggs appear to predominantly express the IP3R1. The three different IP3R subtypes show some differences in sensitivity to InsP3 and Ca2+ [49]. However, the IP3R1 is markedly different from the IP3R2 and IP3R3 in the way it is regulated by ATP. The IP3R1 is stimulated to increase its open probability in response to ATP, and this contrasts with the type 3 IP3R, which requires ~10-fold higher concentrations to be opened by ATP, and the type 2 IP3R, which is not modulated by ATP [49]. Hence, mammalian eggs, that express high levels of IP3R1s, may be sensitive to ATP in a way that is not seen in many other cell types.
The regulation of the IP3R1 by ATP is allosteric and does not involve ATP hydrolysis. In fact the form of ATP that has been shown to stimulate the IP3R1 in frog oocytes is the Mg2+-free form, or ATP4−, which is different from the MgATP2− that provides the energy for ion pumps and other energy-consuming processes [50]. The half-maximal concentration of ATP4− needed to activate Xenopus oocyte IP3Rs is ~270 μM, which is consistent with other studies that suggest a range of 100 to 400 μM of ‘free’ ATP4− for stimulating the IP3R1 [49,50,51]. About 95% of the ATP in the cytosol is bound to Mg2+ as MgATP2−. However, if the total concentration of ATP is around 3.3 mM for mouse eggs [46] and we take the free Mg2+ concentration as 1 mM, which is reported for mammalian cells [52], then the ATP4− concentration in mouse eggs will be ~200 μM (calculated using Maxchelator software [53]). Hence, the ATP4− concentration in mouse eggs is within the range in which it could physiologically modulate the opening of the IP3R1. The idea that ATP4− physiologically modulates the IP3R1 was previously suggested for frog oocytes [50].
We have found that the concentration of total ATP in mouse eggs is higher than in human eggs. Mouse eggs have a total ATP concentration of around 3.3 mM, whereas we found that it was 1.4 mM for human eggs, which is equivalent to about 90 μM of ATP4− [46]. The difference in total ATP concentrations between mouse and human eggs is evident from some previous studies but may have gone unnoticed because of the practice of reporting ATP in picomoles/egg rather than in metric units. Reports state that human eggs have an ATP/egg about twice that of mouse eggs, but the human egg is nearly five times the volume of a mouse egg, so human eggs actually have a lower total ATP concentration [54,55]. Interestingly, data from one study reported that mouse eggs have a total ATP concentration about three times higher than that of hamster eggs [56]. Hence, it is likely that the homeostatic or ‘set concentration’ of total ATP and hence ATP4− may be significantly higher in mouse eggs compared to eggs from many other mammalian species. These differences may not have a large impact on the need for the energetic use of MgATP, for example, in pumping Ca2+ into the ER, because the energy available depends upon the logarithm of the concentration ratio of MgATP over MgADP plus phosphate.
We have found that the concentration of total ATP appears to have a significant effect on Sr2+-induced Ca2+ release in mouse eggs. One can reduce total ATP levels by removing metabolites, such as pyruvate, from the culture medium [57,58]. When mouse eggs are placed in a medium containing Sr2+ but with no pyruvate, they have low ATP and fail to show any Ca2+ oscillations, which is unusual for mouse eggs [46]. When pyruvate is added back to the medium, the ATP levels increase, and Ca2+ oscillations are triggered (see the example in Figure 4) [46]. These data clearly suggest that the concentration of ATP has a significant role in modulating the sensitivity of Ca2+ release in eggs. It is also worth noting that most of the Ca2+ oscillations in these pyruvate-Sr2+ experiments occurred during the rising phase of the ATP (Figure 3) [46]. This may be surprising because there are fewer Ca2+ oscillations after ATP reaches its peak. However, it should be noted that firefly luciferase responds to MgATP and not to ATP4−, which is probably the key modulator of IP3R1s. ATP4− will be produced at the greatest rate during the rising phase of MgATP because mitochondria uniquely produce ATP4−, which they export into the cytosol via the adenine nucleotide exchanger [59]. Since mitochondria are often within 50 nm of the endoplasmic reticulum, it is possible that in responding to ATP4−, the IP3R1 is more sensitive to the activity of mitochondria rather than the concentration of total ATP, even though the two are obviously related (see Figure 3) [23,58].
These studies of ATP in mouse eggs imply that human eggs, and possibly eggs from domesticated animals, could be induced to undergo Ca2+ oscillations in response to a Sr2+ medium if a suitable mechanism could be found to promote mitochondrial activity. Increasing ATP production in eggs is not trivial since adding extra substrates, such as pyruvate, above the level in a standard culture medium, does not increase the ATP concentration [61]. Egg mitochondria may be difficult to stimulate because they may be ‘downregulated’ in their activity as part of a mechanism to reduce reactive oxygen species production, which could otherwise damage mitochondrial DNA [61,62]. However, it is worth noting that that there is an increase in ATP concentration at fertilization in mouse eggs. This is caused in part by the Ca2+ stimulation of mitochondrial dehydrogenases [14,58]. The increase occurs in two phases, and the second phase of increase occurs about 1 h after the start of Ca2+ oscillations [14,57]. The ATP concentration increase is about twofold, so even the mouse egg, with its relatively high ATP concentration, has the capacity to increase the level of ATP given an appropriate stimulus (Figure 4).

7. Conclusions

Many of the studies of Ca2+ release in eggs rely upon research on Ca2+ signaling in somatic cells. I have highlighted important features of Ca2+ release in mammalian eggs that make them different to somatic cells. Eggs are very large spherical cells, and most eggs have a Ca2+ wave at fertilization that propagates throughout the cytoplasm. I suggest that the fast Ca2+ waves at fertilization in mammalian eggs require that PIP2 is distributed throughout the cytoplasm. Interestingly, substantial amounts of PIP2 appear to be associated with yolk granules in the cytoplasm of eggs from frogs and sea urchins [63,64]. The Ca2+ wave in these and other non-mammalian species is not as fast, but some mathematical models of invertebrate eggs suggest that the generation of InsP3 from intracellular sources may also be needed to generate these Ca2+ waves [65].
Another special feature of eggs is that they all appear to express a version of the type 1 IP3R, and eggs from all species contain a high density of mitochondria. There is now evidence that the modulation of IP3R1s can occur by mitochondrially generated ATP4− in mouse eggs. The effects of ATP4− may be a more widespread phenomenon, and it could affect the ability of eggs from different species to generate Ca2+ waves or oscillations in response to sperm at fertilization.

Funding

This research received no external funding.

Acknowledgments

The author thanks Thomas Woolley and Cindy Ikie-Eshalomi for discussions and comments on the manuscript.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Cuthbertson, K.S.; Cobbold, P.H. Phorbol ester and sperm activate mouse oocytes by inducing sustained oscillations in cell Ca2+. Nature 1985, 316, 541–542. [Google Scholar] [CrossRef]
  2. Igusa, Y.; Miyazaki, S. Periodic increase of cytoplasmic free calcium in fertilized hamster eggs measured with calcium-sensitive electrodes. J. Physiol. 1986, 377, 193–205. [Google Scholar] [CrossRef]
  3. Miyazaki, S.; Hashimoto, N.; Yoshimoto, Y.; Kishimoto, T.; Igusa, Y.; Hiramoto, Y. Temporal and spatial dynamics of the periodic increase in intracellular free calcium at fertilization of golden hamster eggs. Dev. Biol. 1986, 118, 259–267. [Google Scholar] [CrossRef] [PubMed]
  4. Kline, D.; Kline, J.T. Repetitive calcium transients and the role of calcium in exocytosis and cell cycle activation in the mouse egg. Dev. Biol. 1992, 149, 80–89. [Google Scholar] [CrossRef] [PubMed]
  5. Miyazaki, S. Intracellular calcium oscillations in mammalian eggs at fertilization. J. Physiol. 2007, 584, 713–714. [Google Scholar] [CrossRef]
  6. Swann, K.; Lai, F.A. Egg Activation at Fertilization by a Soluble Sperm Protein. Physiol. Rev. 2016, 96, 127–149. [Google Scholar] [CrossRef] [PubMed]
  7. Saunders, C.M.; Larman, M.G.; Parrington, J.; Cox, L.J.; Royse, J.; Blayney, L.M.; Swann, K.; Lai, F.A. PLC zeta: A sperm-specific trigger of Ca2+ oscillations in eggs and embryo development. Development 2002, 129, 3533–3544. [Google Scholar] [CrossRef]
  8. Sato, M.S.; Yoshitomo, M.; Mohri, T.; Miyazaki, S. Spatiotemporal analysis of [Ca2+]i rises in mouse eggs after intracytoplasmic sperm injection (ICSI). Cell Calcium 1999, 26, 49–58. [Google Scholar] [CrossRef]
  9. Nozawa, K.; Satouh, Y.; Fujimoto, T.; Oji, A.; Ikawa, M. Sperm-borne phospholipase C zeta-1 ensures monospermic fertilization in mice. Sci. Rep. 2018, 8, 1315. [Google Scholar] [CrossRef]
  10. Stein, P.; Savy, V.; Williams, A.M.; Williams, C.J. Modulators of calcium signalling at fertilization. Open Biol. 2020, 10, 200118. [Google Scholar] [CrossRef] [PubMed]
  11. Wakai, T.; Mehregan, A.; Fissore, R.A. Ca2+ Signaling and Homeostasis in Mammalian Oocytes and Eggs. Cold Spring Harb. Perspect. Biol. 2019, 11, a035162. [Google Scholar] [CrossRef]
  12. Ducibella, T.; Schultz, R.M.; Ozil, J.P. Role of calcium signals in early development. Semin. Cell Dev. Biol. 2006, 17, 324–332. [Google Scholar] [CrossRef]
  13. Jones, K.T. Intracellular calcium in the fertilization and development of mammalian eggs. Clin. Exp. Pharmacol. Physiol. 2007, 34, 1084–1089. [Google Scholar] [CrossRef] [PubMed]
  14. Campbell, K.; Swann, K. Ca2+ oscillations stimulate an ATP increase during fertilization of mouse eggs. Dev. Biol. 2006, 298, 225–233. [Google Scholar] [CrossRef] [PubMed]
  15. Deguchi, R.; Shirakawa, H.; Oda, S.; Mohri, T.; Miyazaki, S. Spatiotemporal analysis of Ca2+ waves in relation to the sperm entry site and animal-vegetal axis during Ca2+ oscillations in fertilized mouse eggs. Dev. Biol. 2000, 218, 299–313. [Google Scholar] [CrossRef]
  16. Swann, K. Soluble sperm factors and Ca2+ release in eggs at fertilization. Rev. Reprod. 1996, 1, 33–39. [Google Scholar] [CrossRef] [PubMed]
  17. Yu, Y.; Nomikos, M.; Theodoridou, M.; Nounesis, G.; Lai, F.A.; Swann, K. PLCζ causes Ca2+ oscillations in mouse eggs by targeting intracellular and not plasma membrane PI(4,5)P2. Mol. Biol. Cell 2012, 23, 371–380. [Google Scholar] [CrossRef] [PubMed]
  18. Sanders, J.R.; Ashley, B.; Moon, A.; Woolley, T.E.; Swann, K. PLCζ Induced Ca2+ Oscillations in Mouse Eggs Involve a Positive Feedback Cycle of Ca2+ Induced InsP3 Formation From Cytoplasmic PIP2. Front. Cell Dev. Biol. 2018, 6, 36. [Google Scholar] [CrossRef]
  19. Sneyd, J.; Tsaneva-Atanasova, K.; Reznikov, V.; Bai, Y.; Sanderson, M.J.; Yule, D.I. A method for determining the dependence of calcium oscillations on inositol trisphosphate oscillations. Proc. Natl. Acad. Sci. USA 2006, 103, 1675–1680. [Google Scholar] [CrossRef]
  20. Politi, A.; Gaspers, L.D.; Thomas, A.P.; Höfer, T. Models of IP3 and Ca2+ oscillations: Frequency encoding and identification of underlying feedbacks. Biophys. J. 2006, 90, 3120–3133. [Google Scholar] [CrossRef]
  21. Miyazaki, S.; Yuzaki, M.; Nakada, K.; Shirakawa, H.; Nakanishi, S.; Nakade, S.; Mikoshiba, K. Block of Ca2+ wave and Ca2+ oscillation by antibody to the inositol 1,4,5-trisphosphate receptor in fertilized hamster eggs. Science 1992, 257, 251–255. [Google Scholar] [CrossRef]
  22. Lee, B.; Yoon, S.Y.; Malcuit, C.; Parys, J.B.; Fissore, R.A. Inositol 1,4,5-trisphosphate receptor 1 degradation in mouse eggs and impact on [Ca2+]i oscillations. J. Cell Physiol. 2010, 222, 238–247. [Google Scholar] [CrossRef] [PubMed]
  23. Foskett, J.K.; White, C.; Cheung, K.H.; Mak, D.O. Inositol trisphosphate receptor Ca2+ release channels. Physiol. Rev. 2007, 87, 593–658. [Google Scholar] [CrossRef] [PubMed]
  24. Kouchi, Z.; Fukami, K.; Shikano, T.; Oda, S.; Nakamura, Y.; Takenawa, T.; Miyazaki, S. Recombinant phospholipase Czeta has high Ca2+ sensitivity and induces Ca2+ oscillations in mouse eggs. J. Biol. Chem. 2004, 279, 10408–10412. [Google Scholar] [CrossRef] [PubMed]
  25. Nomikos, M.; Blayney, L.M.; Larman, M.G.; Campbell, K.; Rossbach, A.; Saunders, C.M.; Swann, K.; Lai, F.A. Role of phospholipase C-zeta domains in Ca2+-dependent phosphatidylinositol 4,5-bisphosphate hydrolysis and cytoplasmic Ca2+ oscillations. J. Biol. Chem. 2005, 280, 31011–31018. [Google Scholar] [CrossRef] [PubMed]
  26. Mak, D.O.; McBride, S.; Foskett, J.K. Inositol 1,4,5-trisphosphate [correction of tris-phosphate] activation of inositol trisphosphate [correction of tris-phosphate] receptor Ca2+ channel by ligand tuning of Ca2+ inhibition. Proc. Natl. Acad. Sci. USA 1998, 95, 15821–15825. [Google Scholar] [CrossRef] [PubMed]
  27. Sienaert, I.; Missiaen, L.; De Smedt, H.; Parys, J.B.; Sipma, H.; Casteels, R. Molecular and functional evidence for multiple Ca2+-binding domains in the type 1 inositol 1,4,5-trisphosphate receptor. J. Biol. Chem. 1997, 272, 25899–25906. [Google Scholar] [CrossRef] [PubMed]
  28. Ornelas-Guevara, R.; Gil, D.; Voorsluijs, V.; Dupont, G. Computational investigation of IP3 diffusion. Sci. Rep. 2023, 13, 2922. [Google Scholar] [CrossRef] [PubMed]
  29. Shirakawa, H.; Ito, M.; Sato, M.; Umezawa, Y.; Miyazaki, S. Measurement of intracellular IP3 during Ca2+ oscillations in mouse eggs with GFP-based FRET probe. Biochem. Biophys. Res. Commun. 2006, 345, 781–788. [Google Scholar] [CrossRef]
  30. Matsu-Ura, T.; Shirakawa, H.; Suzuki, K.G.N.; Miyamoto, A.; Sugiura, K.; Michikawa, T.; Kusumi, A.; Mikoshiba, K. Dual-FRET imaging of IP3 and Ca2+ revealed Ca2+-induced IP3 production maintains long lasting Ca2+ oscillations in fertilized mouse eggs. Sci. Rep. 2019, 9, 4829. [Google Scholar] [CrossRef]
  31. Igusa, Y.; Miyazaki, S. Effects of altered extracellular and intracellular calcium concentration on hyperpolarizing responses of the hamster egg. J. Physiol. 1983, 340, 611–632. [Google Scholar] [CrossRef] [PubMed]
  32. Wakai, T.; Zhang, N.; Vangheluwe, P.; Fissore, R.A. Regulation of endoplasmic reticulum Ca2+ oscillations in mammalian eggs. J. Cell Sci. 2013, 126, 5714–5724. [Google Scholar] [CrossRef] [PubMed]
  33. Bernhardt, M.L.; Stein, P.; Carvacho, I.; Krapp, C.; Ardestani, G.; Mehregan, A.; Umbach, D.M.; Bartolomei, M.S.; Fissore, R.A.; Williams, C.J. TRPM7 and CaV3.2 channels mediate Ca2+ influx required for egg activation at fertilization. Proc. Natl. Acad. Sci. USA 2018, 115, E10370–E10378. [Google Scholar] [CrossRef] [PubMed]
  34. Tsien, R.Y. A non-disruptive technique for loading calcium buffers and indicators into cells. Nature 1981, 290, 527–528. [Google Scholar] [CrossRef] [PubMed]
  35. Hachem, A.; Godwin, J.; Ruas, M.; Lee, H.C.; Ferrer Buitrago, M.; Ardestani, G.; Bassett, A.; Fox, S.; Navarrete, F.; de Sutter, P.; et al. PLCζ is the physiological trigger of the Ca2+ oscillations that induce embryogenesis in mammals but conception can occur in its absence. Development 2017, 144, 2914–2924. [Google Scholar] [CrossRef]
  36. Wu, A.T.; Sutovsky, P.; Manandhar, G.; Xu, W.; Katayama, M.; Day, B.N.; Park, K.W.; Yi, Y.J.; Xi, Y.W.; Prather, R.S.; et al. PAWP, a sperm-specific WW domain-binding protein, promotes meiotic resumption and pronuclear development during fertilization. J. Biol. Chem. 2007, 282, 12164–12175. [Google Scholar] [CrossRef] [PubMed]
  37. Aarabi, M.; Balakier, H.; Bashar, S.; Moskovtsev, S.I.; Sutovsky, P.; Librach, C.L.; Oko, R. Sperm-derived WW domain-binding protein, PAWP, elicits calcium oscillations and oocyte activation in humans and mice. FASEB J. 2014, 28, 4434–4440. [Google Scholar] [CrossRef]
  38. Kang, W.; Harada, Y.; Yamatoya, K.; Kawano, N.; Kanai, S.; Miyamoto, Y.; Nakamura, A.; Miyado, M.; Hayashi, Y.; Kuroki, Y.; et al. Extra-mitochondrial citrate synthase initiates calcium oscillation and suppresses age-dependent sperm dysfunction. Lab. Investig. 2020, 100, 583–595. [Google Scholar] [CrossRef]
  39. Nomikos, M.; Sanders, J.R.; Kashir, J.; Sanusi, R.; Buntwal, L.; Love, D.; Ashley, P.; Sanders, D.; Knaggs, P.; Bunkheila, A.; et al. Functional disparity between human PAWP and PLCζ in the generation of Ca2+ oscillations for oocyte activation. Mol. Hum. Reprod. 2015, 21, 702–710. [Google Scholar] [CrossRef]
  40. Nomikos, M.; Sanders, J.R.; Theodoridou, M.; Kashir, J.; Matthews, E.; Nounesis, G.; Lai, F.A.; Swann, K. Sperm-specific post-acrosomal WW-domain binding protein (PAWP) does not cause Ca2+ release in mouse oocytes. Mol. Hum. Reprod. 2014, 20, 938–947. [Google Scholar] [CrossRef] [PubMed]
  41. Galione, A.; Swann, K.; Georgiou, P.; Whitaker, M. Regenerative and non-regenerative calcium transients in hamster eggs triggered by inositol 1,4,5-trisphosphate. J. Physiol. 1994, 480 Pt 3, 465–474. [Google Scholar] [CrossRef]
  42. Swann, K. Ca2+ oscillations and sensitization of Ca2+ release in unfertilized mouse eggs injected with a sperm factor. Cell Calcium 1994, 15, 331–339. [Google Scholar] [CrossRef] [PubMed]
  43. Yu, Y.; Saunders, C.M.; Lai, F.A.; Swann, K. Preimplantation development of mouse oocytes activated by different levels of human phospholipase C zeta. Hum. Reprod. 2008, 23, 365–373. [Google Scholar] [CrossRef] [PubMed]
  44. Lu, Y.; Reddy, R.; Ferrer Buitrago, M.; Vander Jeught, M.; Neupane, J.; De Vos, W.H.; Van den Abbeel, E.; Lierman, S.; De Sutter, P.; Heindryckx, B. Strontium fails to induce Ca2+ release and activation in human oocytes despite the presence of functional TRPV3 channels. Hum. Reprod. Open 2018, 2018, hoy005. [Google Scholar] [CrossRef]
  45. Tomashov-Matar, R.; Tchetchik, D.; Eldar, A.; Kaplan-Kraicer, R.; Oron, Y.; Shalgi, R. Strontium-induced rat egg activation. Reproduction 2005, 130, 467–474. [Google Scholar] [CrossRef] [PubMed]
  46. Storey, A.; Elgmati, K.; Wang, Y.; Knaggs, P.; Swann, K. The role of ATP in the differential ability of Sr2+ to trigger Ca2+ oscillations in mouse and human eggs. Mol. Hum. Reprod. 2021, 27, gaaa086. [Google Scholar] [CrossRef] [PubMed]
  47. Mann, J.S.; Lowther, K.M.; Mehlmann, L.M. Reorganization of the endoplasmic reticulum and development of Ca2+ release mechanisms during meiotic maturation of human oocytes. Biol. Reprod. 2010, 83, 578–583. [Google Scholar] [CrossRef] [PubMed]
  48. Fissore, R.A.; Longo, F.J.; Anderson, E.; Parys, J.B.; Ducibella, T. Differential distribution of inositol trisphosphate receptor isoforms in mouse oocytes. Biol. Reprod. 1999, 60, 49–57. [Google Scholar] [CrossRef]
  49. Tu, H.; Wang, Z.; Nosyreva, E.; De Smedt, H.; Bezprozvanny, I. Functional characterization of mammalian inositol 1,4,5-trisphosphate receptor isoforms. Biophys. J. 2005, 88, 1046–1055. [Google Scholar] [CrossRef]
  50. Mak, D.O.; McBride, S.; Foskett, J.K. ATP regulation of type 1 inositol 1,4,5-trisphosphate receptor channel gating by allosteric tuning of Ca2+ activation. J. Biol. Chem. 1999, 274, 22231–22237. [Google Scholar] [CrossRef]
  51. Bezprozvanny, I.; Ehrlich, B.E. ATP modulates the function of inositol 1,4,5-trisphosphate-gated channels at two sites. Neuron 1993, 10, 1175–1184. [Google Scholar] [CrossRef] [PubMed]
  52. Konishi, M. Cytoplasmic free concentrations of Ca2+ and Mg2+ in skeletal muscle fibers at rest and during contraction. Jpn. J. Physiol. 1998, 48, 421–438. [Google Scholar] [CrossRef]
  53. Bers, D.M.; Patton, C.W.; Nuccitelli, R. A practical guide to the preparation of Ca2+ buffers. Methods Cell Biol. 1994, 40, 3–29. [Google Scholar] [CrossRef] [PubMed]
  54. Marshall, I.C.; Taylor, C.W. Two calcium-binding sites mediate the interconversion of liver inositol 1,4,5-trisphosphate receptors between three conformational states. Biochem. J. 1994, 301 Pt 2, 591–598. [Google Scholar] [CrossRef]
  55. Van Blerkom, J.; Davis, P.W.; Lee, J. ATP content of human oocytes and developmental potential and outcome after in-vitro fertilization and embryo transfer. Hum. Reprod. 1995, 10, 415–424. [Google Scholar] [CrossRef]
  56. Van Blerkom, J.; Davis, P.; Alexander, S. Inner mitochondrial membrane potential (DeltaPsim), cytoplasmic ATP content and free Ca2+ levels in metaphase II mouse oocytes. Hum. Reprod. 2003, 18, 2429–2440. [Google Scholar] [CrossRef] [PubMed]
  57. Simsek-Duran, F.; Li, F.; Ford, W.; Swanson, R.J.; Jones, H.W., Jr.; Castora, F.J. Age-associated metabolic and morphologic changes in mitochondria of individual mouse and hamster oocytes. PLoS ONE 2013, 8, e64955. [Google Scholar] [CrossRef]
  58. Dumollard, R.; Campbell, K.; Halet, G.; Carroll, J.; Swann, K. Regulation of cytosolic and mitochondrial ATP levels in mouse eggs and zygotes. Dev. Biol. 2008, 316, 431–440. [Google Scholar] [CrossRef]
  59. Dumollard, R.; Marangos, P.; Fitzharris, G.; Swann, K.; Duchen, M.; Carroll, J. Sperm-triggered [Ca2+] oscillations and Ca2+ homeostasis in the mouse egg have an absolute requirement for mitochondrial ATP production. Development 2004, 131, 3057–3067. [Google Scholar] [CrossRef] [PubMed]
  60. Lemasters, J.J. Metabolic implications of non-electrogenic ATP/ADP exchange in cancer cells: A mechanistic basis for the Warburg effect. Biochim. Biophys. Acta Bioenerg. 2021, 1862, 148410. [Google Scholar] [CrossRef]
  61. Dumollard, R.; Carroll, J.; Duchen, M.R.; Campbell, K.; Swann, K. Mitochondrial function and redox state in mammalian embryos. Semin. Cell Dev. Biol. 2009, 20, 346–353. [Google Scholar] [CrossRef] [PubMed]
  62. de Paula, W.B.; Lucas, C.H.; Agip, A.N.; Vizcay-Barrena, G.; Allen, J.F. Energy, ageing, fidelity and sex: Oocyte mitochondrial DNA as a protected genetic template. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2013, 368, 20120263. [Google Scholar] [CrossRef] [PubMed]
  63. Snow, P.; Yim, D.L.; Leibow, J.D.; Saini, S.; Nuccitelli, R. Fertilization stimulates an increase in inositol trisphosphate and inositol lipid levels in Xenopus eggs. Dev. Biol. 1996, 180, 108–118. [Google Scholar] [CrossRef] [PubMed]
  64. Rice, A.; Parrington, J.; Jones, K.T.; Swann, K. Mammalian sperm contain a Ca2+-sensitive phospholipase C activity that can generate InsP3 from PIP2 associated with intracellular organelles. Dev. Biol. 2000, 228, 125–135. [Google Scholar] [CrossRef]
  65. Dupont, G.; Dumollard, R. Simulation of calcium waves in ascidian eggs: Insights into the origin of the pacemaker sites and the possible nature of the sperm factor. J. Cell Sci. 2004, 117, 4313–4323. [Google Scholar] [CrossRef]
Figure 2. A schematic illustration of the changing pattern of Ca2+ waves in mouse and hamster eggs at fertilization. The initial Ca2+ increase is triggered by PLCζ in a restricted region of the egg near the site of sperm–egg fusion. After about 10–15 min, PLCζ will have diffused throughout the egg cytoplasm. The very fast waves are now driven by Ca2+-induced InsP3 formation from cytoplasmic PIP2.
Figure 2. A schematic illustration of the changing pattern of Ca2+ waves in mouse and hamster eggs at fertilization. The initial Ca2+ increase is triggered by PLCζ in a restricted region of the egg near the site of sperm–egg fusion. After about 10–15 min, PLCζ will have diffused throughout the egg cytoplasm. The very fast waves are now driven by Ca2+-induced InsP3 formation from cytoplasmic PIP2.
Cells 12 02809 g002
Figure 3. A schematic diagram of the way ATP4− may regulate the IP3R1 in eggs. The IP3R1 in the ER membrane can respond to Ca2+ at two sites, one inhibitory and one stimulatory. InsP3 binding inhibits the inhibitory site (−ve) to stimulate Ca2+ release [26,60]. ATP4− increases the Ca2+ sensitivity of the stimulatory site (+ve) to promote Ca2+ release [50]. Sr2+ appears to promote IP3R1 opening by binding predominantly to the stimulatory site [27,60]. Mitochondria export ATP into the cytosol in the form of ATP4−, which will rapidly form a complex with Mg2+, but ATP4− may be more concentrated in regions close to the mitochondrial outer membrane.
Figure 3. A schematic diagram of the way ATP4− may regulate the IP3R1 in eggs. The IP3R1 in the ER membrane can respond to Ca2+ at two sites, one inhibitory and one stimulatory. InsP3 binding inhibits the inhibitory site (−ve) to stimulate Ca2+ release [26,60]. ATP4− increases the Ca2+ sensitivity of the stimulatory site (+ve) to promote Ca2+ release [50]. Sr2+ appears to promote IP3R1 opening by binding predominantly to the stimulatory site [27,60]. Mitochondria export ATP into the cytosol in the form of ATP4−, which will rapidly form a complex with Mg2+, but ATP4− may be more concentrated in regions close to the mitochondrial outer membrane.
Cells 12 02809 g003
Figure 4. A recording of Ca2+ and ATP changes in mouse eggs incubated in a medium containing 5 mM Sr2+, as described and presented previously in Storey et al. [46]. The Ca2+ changes are measured with a fluorescent dye and shown in red; the ATP changes are monitored using the luminescence of luciferase and shown in black. In the initial part of the recording, the eggs are in a medium that lacks any metabolites; it has low ATP, and no Ca2+ oscillations are seen. After ~40 min, pyruvate (1 mM) is added to the medium containing the eggs. This leads to an increase in ATP and the triggering of Ca2+ oscillations.
Figure 4. A recording of Ca2+ and ATP changes in mouse eggs incubated in a medium containing 5 mM Sr2+, as described and presented previously in Storey et al. [46]. The Ca2+ changes are measured with a fluorescent dye and shown in red; the ATP changes are monitored using the luminescence of luciferase and shown in black. In the initial part of the recording, the eggs are in a medium that lacks any metabolites; it has low ATP, and no Ca2+ oscillations are seen. After ~40 min, pyruvate (1 mM) is added to the medium containing the eggs. This leads to an increase in ATP and the triggering of Ca2+ oscillations.
Cells 12 02809 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Swann, K. Sperm-Induced Ca2+ Release in Mammalian Eggs: The Roles of PLCζ, InsP3, and ATP. Cells 2023, 12, 2809. https://doi.org/10.3390/cells12242809

AMA Style

Swann K. Sperm-Induced Ca2+ Release in Mammalian Eggs: The Roles of PLCζ, InsP3, and ATP. Cells. 2023; 12(24):2809. https://doi.org/10.3390/cells12242809

Chicago/Turabian Style

Swann, Karl. 2023. "Sperm-Induced Ca2+ Release in Mammalian Eggs: The Roles of PLCζ, InsP3, and ATP" Cells 12, no. 24: 2809. https://doi.org/10.3390/cells12242809

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop