Volume 6, Issue 5 (7-2008)                   IJRM 2008, 6(5): 171-174 | Back to browse issues page

XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Heytens E, Soleimani R, De Sutter P. Oocyte activation: The impact of calcium signals on fertilization. IJRM 2008; 6 (5) :171-174
URL: http://ijrm.ir/article-1-127-en.html
1- Department of Reproductive Medicine, University Hospital Ghent, Ghent, Belgium , elke.heytens@ugent.be
2- Department of Reproductive Medicine, University Hospital Ghent, Ghent, Belgium
Full-Text [PDF 169 kb]   (973 Downloads)     |   Abstract (HTML)  (2060 Views)
Full-Text:   (318 Views)
     Introduction
Calcium is a universal secondary messenger in cells controlling diverse biological processes such as proliferation, differentiation, axis formation, transcriptional activation and apoptosis (1). It plays a major role at fertilization and is thus already involved at the very beginning of life. Sperm not only delivers its genetic material but also triggers rises in intracellular calcium concentration ([Ca2+]i) and consequently awakens the oocyte which is blocked at the metaphase of the second meiotic division (MII arrest). For all species an increase in [Ca2+]i is necessary and sufficient for the completion of oocyte activation and initiation of embryonic development (2,3). In mammalian oocytes, a series of spikes  in  [ Ca2+ ] i  or so called  Ca2+-oscillations, are triggered upon sperm entry. These  Ca2+- oscillations  are needed to relieve MII arrest and to provoke all the other events of oocyte activation, such as the  cortical  reaction ,  maternal mRNA recruitment, pronuclear development and mitotic cleavage ( 4). MII arrest is characterized by a high level of the cyclinB/cdk1 complex, also known as maturation promoting factor (MPF). MPF prompts metaphase II and is thought to be involved in several central features of cell division, such as disassembly of the nucleus, chromosome condensation, cytoskeletal rearrangements and arrest in transcriptional activity (5).
     MPF activity in the MII-arrested oocyte is regulated by the activity of a cytostatic factor (CSF). During MII arrest, CSF indeed prevents the destruction of MPF by keeping the anaphase-promoting complex (APC) inactive (figure 1). When active, the latter complex labels cyclin B for degradation through the ubiquitin pathway. At fertilization, MPF levels must decrease sufficiently to enable the oocyte to complete the meiosis and initiate oocyte activation (6).
     Several theories on how the sperm sparks Ca2+-oscillations and thus activates the oocyte have been proposed. In the surface-mediated model of oocyte activation, binding of a sperm protein on a putative oocyte receptor would activate PLC which in turn would produce inositol 1,4,5 trisphosphate, which induces release of Ca2+ from intracellular stores and subsequently triggers Ca2+-oscillations in the oocyte (2). However, although in echinoderms and amphibians there is evidence of involvement of such an oocyte PLC, no such evidence has been forthcoming for mammals and in general there is a lack of evidence showing that a surface protein that mediates sperm-oocyte binding is linked with intracellular signalling leading to oocyte activation (7). A second model in which a soluble sperm factor introduced into the ooplasm at gamete fusion activates the oocyte was proposed by Dale et al, on the basis that microinjection of sea urchin sperm extract triggered the cortical reaction in sea urchin eggs (8). Subsequently Swann and others showed that injection of mammalian sperm extracts into eggs and oocytes resulted in a pattern of Ca2+ release similar to that seen at fertilization and also triggered egg activation (9, 10).
 
The oocyte activating factor PLCζ
     The identity of the sperm factor remained unknown for some time, but then a sperm specific phospholipase C named PLCζ was identified which had the expected properties of the oocyte activating factor (11). Injection of recombinant mouse and human PLCζ RNA into mouse oocytes not only evokes Ca2+-oscillations similar to those induced by sperm but also promotes subsequent embryo development (11,12).
     A similar response was obtained by microinjecting recombinant mouse PLCζ  protein into mouse oocytes (13). Immunolocalization studies have shown that the pattern of localization of PLCζ in sperm of both rodents and humans is in the expected location for an oocyte activation factor, namely the perinuclear theca (14,15). Moreover, sperm from transgenic mice expressing short hairpin RNAs targeting PLCζ exhibited reduced amounts of PLCζ, and when injected into mouse oocytes, induced Ca2+ oscillations that ended prematurely (16).
     PLCζ catalyzes the formation of inositol trisphosphate, which triggers the release of Ca2+ from intracellular stores and subsequently induces the calcium oscillations in the oocyte (2). Calcium will activate calmodulin dependent kinase II (CaMKII), an enzyme known to be sensitive to the frequency of calcium oscillations (17). CaMKII directly activates APC and also promotes the degradation of CSF (18).
      As a result, the APC is liberated from its repression by CSF and the inhibition of MPF will be relieved. APC activation causes a decline in MPF activity and the concomitant release from the MII arrest (Figure 1).
     The Ca2+-oscillations prevent the MPF levels to return to active levels and bring about successful activation (4).
 
Oocyte activation in human assisted reproduction technology
     Calcium oscillations are also observed after intracytoplasmic sperm injection (ICSI), a highly successful technique used in human assisted reproductive medicine in case of male infertility (19). Only in a small percentage of the patients failed fertilization is observed after ICSI (20). A specific group of patients who often face failed fertilization are men with globozoospermia, a rare disorder characterized by round-headed, acrosome-less sperm cells (21). The lack of an acrosome was considered to be the cause of infertility in these patients (22).
     The introduction of ICSI led to several successful attempts resulting in  pregnancies following ICSI with globozoospermia sperm, however fertilization rates were low (23). Moreover, there have been several reports of unsuccessful ICSI attempts in cases of globozoospermia (24).
     This suggests that besides the inability to interact with the female gamete due to an abnormal acrosome, globozoospermia sperm may have additional defects that affect their fertilization capability. One study employing the mouse oocyte activation test (MOAT) to analyse spermatozoa from globozoospermic men indicated that they could not successfully activate oocytes (25).
     Currently the presence of PLCζ in sperm cells is under investigation and preliminary data confirm that sperm from globozoospermic men contain lower amounts of PLCζ as compared to normal (26). In addition, also other cases of failed fertilization may be explained by defects in the PLCζ protein.
 
Assisted oocyte activation
     Before considering gamete donation, artificial activation of the oocytes may be proposed as an option to couples who experienced repeated cycles with failed fertilization after ICSI. Mammalian oocytes such as murine, rabbit, bovine, rat, porcine and also human oocytes can namely be activated just by increasing [Ca2+]i using electrical activation, ethanol treatment or Ca2+ ionophore challenge (27-32).
     Interestingly, this technique of artificially induced oocyte activation at the time of ICSI or assisted oocyte activation (AOA) by using for example calcium ionophore to increase [Ca2+]i can restore normal fertilization rates in these cases and support further development as evidenced by successful pregnancies and deliveries reported (33-35). However, some concerns still exist about the use of calcium ionophore as an activating agent in human ART (33,35). And although no adverse effects of ionomycin on in vitro or in vivo mouse embryo development were noticed (36), a negative long-term effect of this compound can not be excluded at present. In addition, some studies have shown that the Ca2+-transients induced during oocyte activation may influence the number of embryos reaching the blastocyst stage and even postnatal development (37,38). Ionomycin induces only a single Ca2+ transient in human oocytes (39) and no Ca2+-oscillations are triggered after exposing mouse oocytes injected with human sperm deficient in oocyte activating capacity (40). Successful clinical application in the human, however, suggests that this does not seem to prevent further embryonic development.
     In attempt to induce calcium changes in more physiological way, other techniques have been introduced such as a modified ICSI technique (41) and recently also strontium was applied successfully in a clinical setting (42) . Strontium acts like a surrogate of Ca2+ in mouse oocytes and causes a series of Ca2+ -oscillations similar to those observed at fertilization  (28,43). However, the observation that no Ca2+-oscillations were triggered in human oocytes treated with strontium  (12) suggests that other mechanisms than described for mouse oocytes are involved (43) and that an activation protocol based on strontium should still be treated with caution. Interestingly, the discovery of the oocyte activating factor PLCζ (11) and the observation of PLCζ deficiencies in infertile men (26), may lead to the development of a more physiological method of triggering oocyte activation in case of sperm borne fertilization failure after ICSI, for example by co-injecting a recombinant PLCζ at the time of ICSI.
     Unraveling the mechanism of oocyte activation and determining the role of calcium in this process has resulted in promising applications in human ART. However, follow-up of the children born after assisted oocyte activation remains necessary, whatever protocol is used.



 
Type of Study: Original Article |

References
1. Berridge MJ, Lipp P, Bootman MD. The versatility and universality of calcium signalling. Nat Rev Mol Cell Biol 2000; 1: 11-21. [DOI:10.1038/35036035]
2. Miyazaki S, Shirakawa H, Nakada K, Honda Y. Essential role of the inositol 1,4,5-trisphosphate receptor/Ca2+ release channel in Ca2+ waves and Ca2+ oscillations at fertilization of mammalian eggs. Dev Biol 1993; 158: 62-78. [DOI:10.1006/dbio.1993.1168]
3. Cuthbertson KS, Whittingham DG, Cobbold PH. Free Ca2+ increases in exponential phases during mouse oocyte activation. Nature 1981; 294: 754-757. [DOI:10.1038/294754a0]
4. Ducibella T, Huneau D, Angelichio E, Xu Z, Schultz RM, Kopf GS, et al. Egg-to-embryo transition is driven by differential responses to Ca(2+) oscillation number. Dev Biol 2002;250: 280-291. [DOI:10.1006/dbio.2002.0788]
5. Moreno S , Nurse P. Substrates for p34cdc2: in vivo veritas? Cell 1990; 61: 549-551. [DOI:10.1016/0092-8674(90)90463-O]
6. Murray AW, Solomon MJ , Kirschner MW. The role of cyclin synthesis and degradation in the control of maturation promoting factor activity. Nature 1989; 339: 280-286. [DOI:10.1038/339280a0]
7. Miyazaki S. Thirty years of calcium signals at fertilization. Seminars in Cell and Developmental Biology 2006; 17: 233-243. [DOI:10.1016/j.semcdb.2006.02.007]
8. Dale B, DeFelice LJ , Ehrenstein G. Injection of a soluble sperm fraction into sea-urchin eggs triggers the cortical reaction. Experientia 1985; 41: 1068-1070. [DOI:10.1007/BF01952148]
9. Swann K. A cytosolic sperm factor stimulates repetitive calcium increases and mimics fertilization in hamster eggs. Development 1990; 110: 1295-1302.
10. Dozortsev D, Rybouchkin A, De Sutter P, Qian C ,Dhont M. Human oocyte activation following intracytoplasmic injection: the role of the sperm cell. Hum Reprod 1995; 10: 403-407. [DOI:10.1093/oxfordjournals.humrep.a135952]
11. Saunders CM, Larman MG, Parrington J, Cox LJ, Royse J, Blayney LM, et al. PLC zeta: a sperm-specific trigger of Ca(2+) oscillations in eggs and embryo development. Development 2002; 129: 3533-3544.
12. Rogers NT, Hobson E, Pickering S, Lai FA, Braude P , Swann K. Phospholipase Czeta causes Ca2+ oscillations and parthenogenetic activation of human oocytes. Reproduction 2004;128: 697-702. [DOI:10.1530/rep.1.00484]
13. Kouchi Z, Fukami K, Shikano T, Oda S, Nakamura Y, Takenawa T, et al. Recombinant phospholipase Czeta has high Ca2+ sensitivity and induces Ca2+ oscillations in mouse eggs. J Biol Chem 2004; 279: 10408-10412. [DOI:10.1074/jbc.M313801200]
14. Young C, Grasa P, Coward K, Davis LC , Parrington J. Phospholipase C zeta undergoes dynamic changes in its pattern of localization in sperm during capacitation and the acrosome reaction. Fertil Steril 2008; E-pub ahead of print. [DOI:10.1016/j.fertnstert.2008.05.021]
15. Grasa P, Coward K, Young C , Parrington J. The pattern of localization of the putative egg activation factor, PLCz, in uncapacitated, capacitated, and acrosome-reacted human spermatozoa. Hum Reprod 2008; 23: 2513-2522. [DOI:10.1093/humrep/den280]
16. Knott JG, Kurokawa M, Fissore RA, Schultz RM ,Williams CJ. Transgenic RNA interference reveals role for mouse sperm phospholipase Czeta in triggering Ca2+ oscillations during fertilization. Biol Reprod 2005; 72: 992-996. [DOI:10.1095/biolreprod.104.036244]
17. Dupont G , Goldbeter A. CaM kinase II as frequency decoder of Ca2+ oscillations. Bioessays 1998; 20: 607-610. https://doi.org/10.1002/(SICI)1521-1878(199808)20:8<607::AID-BIES2>3.0.CO;2-F [DOI:10.1002/(SICI)1521-1878(199808)20:83.0.CO;2-F]
18. Madgwick S, Levasseur M , Jones KT. Calmodulin-dependent protein kinase II, and not protein kinase C, is sufficient for triggering cell-cycle resumption in mammalian eggs. J Cell Sci 2005; 118: 3849-3859. [DOI:10.1242/jcs.02506]
19. Tesarik J, Sousa M , Testart J. Human oocyte activation after intracytoplasmic sperm injection. Hum Reprod 1994; 9: 511-518. [DOI:10.1093/oxfordjournals.humrep.a138537]
20. Sousa M , Tesarik J. Ultrastructural analysis of fertilization failure after intracytoplasmic sperm injection. Hum Reprod 1994; 9: 2374-2380. [DOI:10.1093/oxfordjournals.humrep.a138455]
21. Schirren CG, Holstein A , Schirren C. Uber die Morphogenese rundkopfiger Spermatozoen des Menschen. Andrologie 1971;3: 117-125. [DOI:10.1111/j.1439-0272.1971.tb02106.x]
22. Holstein AF, Schirren CG, Schirren C , Mauss J. Round headed spermatozoa: a cause of male infertility. Dtsch Med Wochenschr 1973; 98: 61-62. [DOI:10.1055/s-0028-1106747]
23. Liu J, Nagy Z, Joris H, Tournaye H, Devroey P, Van Steirteghem A. Successful fertilization and establishment of pregnancies after intracytoplasmic sperm injection in patients with globozoospermia. Hum Reprod 1995; 10: 626-629. [DOI:10.1093/oxfordjournals.humrep.a136000]
24. Battaglia DE, Koehler JK, Klein NA, Tucker MJ. Failure of oocyte activation after intracytoplasmic sperm injection using round-headed sperm. Fertil Steril 1997; 68: 118-122. [DOI:10.1016/S0015-0282(97)81486-0]
25. Rybouchkin A, Dozortsev D, Pelinck MJ, De Sutter P, Dhont M. Analysis of the oocyte activating capacity and chromosomal complement of round-headed human spermatozoa by their injection into mouse oocytes. Hum Reprod 1996; 11: 2170-2175. [DOI:10.1093/oxfordjournals.humrep.a019071]
26. Heytens E, Parrington J, Lambrecht S, Young C, Coward K, Cuvelier C, et al. First evidence of disturbed expression of the oocyte-activating factor PLCzeta in globozoospermic men. Hum Reprod 2008; 23: i103.
27. Ware CB, Barnes FL, Maiki-Laurila M , First NL. Age dependence of bovine oocyte activation. Gamete Res 1989; 22: 265-275. [DOI:10.1002/mrd.1120220304]
28. Kline D , Kline JT. Repetitive calcium transients and the role of calcium in exocytosis and cell cycle activation in the mouse egg. Dev Biol 1992; 149: 80-89. [DOI:10.1016/0012-1606(92)90265-I]
29. Im GS, Seo JS, Hwang IS, Kim DH, Kim SW, Yang BC, et al. Development and apoptosis of pre-implantation porcine nuclear transfer embryos activated with different combination of chemicals. Mol Reprod Dev 2006; 73: 1094-1101. [DOI:10.1002/mrd.20455]
30. Mizutani E, Jiang JY, Mizuno S, Tomioka I, Shinozawa T, Kobayashi J, et al. Determination of optimal conditions for parthenogenetic activation and subsequent development of rat oocytes in vitro. J Reprod Dev 2004; 50: 139-146. [DOI:10.1262/jrd.50.139]
31. Collas P, Fissore R, Robl JM, Sullivan EJ , Barnes FL. Electrically induced calcium elevation, activation, and parthenogenetic development of bovine oocytes. Mol Reprod Dev 1993; 34: 212-223. [DOI:10.1002/mrd.1080340214]
32. Ozil JP. The parthenogenetic development of rabbit oocytes after repetitive pulsatile electrical stimulation. Development 1990; 109: 117-127.
33. Rybouchkin AV, Van der Straeten F, Quatacker J, De Sutter P , Dhont M. Fertilization and pregnancy after assisted oocyte activation and intracytoplasmic sperm injection in a case of round-headed sperm associated with deficient oocyte activation capacity. Fertil Steril 1997; 68: 1144-1147. [DOI:10.1016/S0015-0282(97)00378-6]
34. Heindryckx B, Van der Elst J, De Sutter P, Dhont M. Treatment option for sperm- or oocyte-related fertilization failure: assisted oocyte activation following diagnostic heterologous ICSI. Hum Reprod 2005; 20: 2237-2241. [DOI:10.1093/humrep/dei029]
35. Nasr-Esfahani MH, Razavi S, Javdan Z , Tavalaee M. Artificial oocyte activation in severe teratozoospermia undergoing intracytoplasmic sperm injection. Fertil Steril 2008; 90: 2231-2237. [DOI:10.1016/j.fertnstert.2007.10.047]
36. Heytens E, Soleimani R, Lierman S, De Meester S, Gerris J, Dhont M, et al. Effect of ionomycin on oocyte activation and embryo development in mouse. Reprod Biomed Online 2008; 17: 764-771. [DOI:10.1016/S1472-6483(10)60403-8]
37. Ozil JP, Banrezes B, Toth S, Pan H ,Schultz RM. Ca2+ oscillatory pattern in fertilized mouse eggs affects gene expression and development to term. Dev Biol 2006; 300: 534-544. [DOI:10.1016/j.ydbio.2006.08.041]
38. Ozil JP, Huneau D. Activation of rabbit oocytes: the impact of the Ca2+ signal regime on development. Development 2001; 128: 917-928.
39. Rinaudo P, Pepperell JR, Buradgunta S, Massobrio M, Keefe DL. Dissociation between intracellular calcium elevation and development of human oocytes treated with calcium ionophore. Fertil Steril 1997; 68: 1086-1092. [DOI:10.1016/S0015-0282(97)00406-8]
40. Heytens E, Vanhoutte L, Lierman S, Cuvelier C, Gerris J, Dhont M, et al. Ionophore treatment of mouse oocytes injected with human round-headed sperm does not lead to subsequent normal calcium oscillation pattern. Hum Reprod 2007; 22: i214-i215.
41. Tesarik J, Rienzi L, Ubaldi F, Mendoza C , Greco E. Use of a modified intracytoplasmic sperm injection technique to overcome sperm-borne and oocyte-borne oocyte activation failures. Fertil Steril 2002; 78: 619-624. [DOI:10.1016/S0015-0282(02)03291-0]
42. Yanagida K, Morozumi K, Katayose H, Hayashi S ,Sato A. Successful pregnancy after ICSI with strontium oocyte activation in low rates of fertilization. Reprod Biomed Online 2006; 13: 801-806. [DOI:10.1016/S1472-6483(10)61027-9]
43. Zhang D, Pan L, Yang LH, He XK, Huang XY , Sun FZ. Strontium promotes calcium oscillations in mouse meiotic oocytes and early embryos through InsP3 receptors, and requires activation of phospholipase and the synergistic action of InsP3. Hum Reprod 2005; 20: 3053-3061. [DOI:10.1093/humrep/dei215]

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

Designed & Developed by : Yektaweb