1
|
Abdulsamad HMR, Murtaza ZF, AlMuhairi HM, Bafleh WS, AlMansoori SA, AlQubaisi SA, Hamdan H, Kashir J. The Therapeutic and Diagnostic Potential of Phospholipase C Zeta, Oocyte Activation, and Calcium in Treating Human Infertility. Pharmaceuticals (Basel) 2023; 16:441. [PMID: 36986540 PMCID: PMC10056371 DOI: 10.3390/ph16030441] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/19/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Oocyte activation, a fundamental event during mammalian fertilisation, is initiated by concerted intracellular patterns of calcium (Ca2+) release, termed Ca2+ oscillations, predominantly driven by testis-specific phospholipase C zeta (PLCζ). Ca2+ exerts a pivotal role in not just regulating oocyte activation and driving fertilisation, but also in influencing the quality of embryogenesis. In humans, a failure of Ca2+ release, or defects in related mechanisms, have been reported to result in infertility. Furthermore, mutations in the PLCζ gene and abnormalities in sperm PLCζ protein and RNA, have been strongly associated with forms of male infertility where oocyte activation is deficient. Concurrently, specific patterns and profiles of PLCζ in human sperm have been linked to parameters of semen quality, suggesting the potential for PLCζ as a powerful target for both therapeutics and diagnostics of human fertility. However, further to PLCζ and given the strong role played by Ca2+ in fertilisation, targets down- and up-stream of this process may also present a significantly similar level of promise. Herein, we systematically summarise recent advancements and controversies in the field to update expanding clinical associations between Ca2+-release, PLCζ, oocyte activation and human fertility. We discuss how such associations may potentially underlie defective embryogenesis and recurrent implantation failure following fertility treatments, alongside potential diagnostic and therapeutic avenues presented by oocyte activation for the diagnosis and treatment of human infertility.
Collapse
Affiliation(s)
- Haia M. R. Abdulsamad
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Zoha F. Murtaza
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Hessa M. AlMuhairi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Wjdan S. Bafleh
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Salma A. AlMansoori
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Shaikha A. AlQubaisi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Junaid Kashir
- Department of Biology, College of Arts and Science, Khalifa University, Abu Dhabi 127788, United Arab Emirates
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 12713, Saudi Arabia
| |
Collapse
|
2
|
Wang T, Cao B, Cai Y, Chen S, Wang B, Yuan Y, Zhang Q. Plcz1 Deficiency Decreased Fertility in Male Mice Which Is Associated with Sperm Quality Decline and Abnormal Cytoskeleton in Epididymis. Int J Mol Sci 2022; 24:314. [PMID: 36613757 PMCID: PMC9820195 DOI: 10.3390/ijms24010314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Phospholipase C zeta1 (Plcz1) was known to be a physiological factor in sperm that activates oocytes to complete meiosis by triggering Ca2+ oscillations after fertilisation. However, the role of male Plcz1 in spermatogenesis and early embryo development in progeny has been controversial. Plcz1 knockout (Plcz1-/-) mouse model (Plcz1m3 and Plcz1m5) was generated by using the CRISPR-Cas9 system. The fertility of Plcz1-/- mice was evaluated by analysing the number of offsprings, sperm quality, pathological changes in the testis and epididymis. RNA-seq and RT-PCR were performed to screen differentially expressed genes and signalling pathways related to fertility in Plcz1-/- mice. Further mechanism was explored by using Plcz1-/- cells. Plcz1 knockout led to hypofertility in male mice. In particular, a significant time delay in development and polyspermy was found in eggs fertilized by both Plcz1m3 and Plcz1m5 sperm. Interestingly, a decline in sperm quality combined with pathological changes in epididymis was found in Plcz1m3 mice but not in Plcz1m5 mice. Notably, abnormal cytoskeleton appears in epididymis of Plcz1m3 mice and Plcz1-/- cells. Cytoskeleton damage of epididymis is involved in fertility decline of males upon Plcz1 deficiency in this model.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225012, China
| | - Binbin Cao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225012, China
| | - Yao Cai
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225012, China
| | - Si Chen
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225012, China
| | - Baozhu Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225012, China
| | - Yan Yuan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225012, China
| | - Quan Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225012, China
| |
Collapse
|
3
|
Zafar MI, Lu S, Li H. Sperm-oocyte interplay: an overview of spermatozoon's role in oocyte activation and current perspectives in diagnosis and fertility treatment. Cell Biosci 2021; 11:4. [PMID: 33407934 PMCID: PMC7789549 DOI: 10.1186/s13578-020-00520-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
The fertilizing spermatozoon is a highly specialized cell that selects from millions along the female tract until the oocyte. The paternal components influence the oocyte activation during fertilization and are fundamental for normal embryo development; however, the sperm-oocyte interplay is in a continuous debate. This review aims to analyze the available scientific information related to the role of the male gamete in the oocyte activation during fertilization, the process of the interaction of sperm factors with oocyte machinery, and the implications of any alterations in this interplay, as well as the advances and limitations of the reproductive techniques and diagnostic tests. At present, both PLCζ and PAWP are the main candidates as oocyte activated factors during fertilization. While PLCζ mechanism is via IP3, how PAWP activates the oocyte still no clear, and these findings are important to study and treat fertilization failure due to oocyte activation, especially when one of the causes is the deficiency of PLCζ in the sperm. However, no diagnostic test has been developed to establish the amount of PLCζ, the protocol to treat this type of pathologies is broad, including treatment with ionophores, sperm selection improvement, and microinjection with PLCζ protein or RNA.
Collapse
Affiliation(s)
- Mohammad Ishraq Zafar
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, People's Republic of China
| | - Shi Lu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jeifang Avenue, Wuhan, 430022, People's Republic of China
| | - Honggang Li
- Institute of Reproductive Health/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, People's Republic of China. .,Wuhan Tongji Reproductive Medicine Hospital, 128 Sanyang Road, Wuhan, 430013, People's Republic of China.
| |
Collapse
|
4
|
Kashir J, Mistry BV, Gumssani MA, Rajab M, Abu-Dawas R, AlMohanna F, Nomikos M, Jones C, Abu-Dawud R, Al-Yacoub N, Coward K, Lai FA, Assiri AM. Advancing male age differentially alters levels and localization patterns of PLCzeta in sperm and testes from different mouse strains. Asian J Androl 2021; 23:178-187. [PMID: 33208563 PMCID: PMC7991809 DOI: 10.4103/aja.aja_67_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Sperm-specific phospholipase C zeta (PLCζ) initiates intracellular calcium (Ca2+) transients which drive a series of concurrent events collectively termed oocyte activation. Numerous investigations have linked abrogation and absence/reduction of PLCζ with forms of male infertility in humans where oocyte activation fails. However, very few studies have examined potential relationships between PLCζ and advancing male age, both of which are increasingly considered to be major effectors of male fertility. Initial efforts in humans may be hindered by inherent PLCζ variability within the human population, alongside a lack of sufficient controllable repeats. Herein, utilizing immunoblotting, immunofluorescence, and quantitative reverse transcription PCR (qRT-PCR) we examined for the first time PLCζ protein levels and localization patterns in sperm, and PLCζ mRNA levels within testes, from mice at 8 weeks, 12 weeks, 24 weeks, and 36 weeks of age, from two separate strains of mice, C57BL/6 (B6; inbred) and CD1 (outbred). Collectively, advancing male age generally diminished levels and variability of PLCζ protein and mRNA in sperm and testes, respectively, when both strains were examined. Furthermore, advancing male age altered the predominant pattern of PLCζ localization in mouse sperm, with younger mice exhibiting predominantly post-acrosomal, and older mice exhibiting both post-acrosomal and acrosomal populations of PLCζ. However, the specific pattern of such decline in levels of protein and mRNA was strain-specific. Collectively, our results demonstrate a negative relationship between advancing male age and PLCζ levels and localization patterns, indicating that aging male mice from different strains may serve as useful models to investigate PLCζ in cases of male infertility and subfertility in humans.
Collapse
Affiliation(s)
- Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Bhavesh V Mistry
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Maha Adel Gumssani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Muhammad Rajab
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Reema Abu-Dawas
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Falah AlMohanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha PO Box 2713, Qatar
| | - Celine Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - Raed Abu-Dawud
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Nadya Al-Yacoub
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Kevin Coward
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - F Anthony Lai
- College of Medicine, QU Health, Qatar University, Doha PO Box 2713, Qatar.,Biomedical Research Centre, Qatar University, Doha PO Box 2713, Qatar
| | - Abdullah M Assiri
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
5
|
Soriano-Úbeda C, Avilés-López K, García-Vázquez FA, Romero-Aguirregomezcorta J, Matás C. Epididymal and ejaculated sperm functionality is regulated differently by periovulatory oviductal fluid in pigs. Andrology 2020; 9:426-439. [PMID: 32920990 DOI: 10.1111/andr.12902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND The current results of in vitro reproduction techniques in pigs, such as in vitro fertilization (IVF) and embryo development, show high performance with both epididymal and ejaculated spermatozoa. However, the results using ejaculated spermatozoa are even better. Ejaculated spermatozoa are exposed to the secretions of the accessory seminal glands: the seminal plasma (SP). It has been reported that exposure of spermatozoa to reproductive fluids, such as SP or periovulatory oviductal fluid (pOF), modulates sperm functionality both in vivo and in vitro. But whether or not this modulating effect of pOF depends on the origin of the spermatozoa being epididymal or ejaculated, is still unknown. OBJECTIVES To determine and compare the effect of pOF on epididymal and ejaculated sperm functionality. MATERIAL AND METHODS The effects of incubating spermatozoa from the epididymis and ejaculate with pOF in capacitating conditions were investigated by analyzing sperm motility, phosphorylation of protein kinase A substrates and proteins in tyrosine (pPKAs and pTyr, respectively), the interaction of the spermatozoa with the oocyte in IVF and intracytoplasmic sperm injection (ICSI), and, finally, the spermatozoa chromatin condensation status. RESULTS The pOF modified events related to capacitation in epididymal spermatozoa by decreasing motility, pPKAs and pTyr. In the interaction with the oocyte after sperm capacitation, pOF regulated the epididymal and ejaculated spermatozoa differently. While pOF decreased the number of spermatozoa bound to the zona pellucida (Spz/ZP) and increased oocyte activation after ICSI with epididymal spermatozoa, with the ejaculated spermatozoa, it decreased the mean number penetrating each oocyte (Spz/O). Additionally, pOF significantly increased the nuclear decondensation of the epididymal spermatozoa after the fertilization of the oocyte. CONCLUSION The modulation of sperm functionality by pOF is conditioned by the origin of the spermatozoa.
Collapse
Affiliation(s)
- Cristina Soriano-Úbeda
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Karen Avilés-López
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Francisco A García-Vázquez
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Jon Romero-Aguirregomezcorta
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain.,Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
| | - Carmen Matás
- Department of Physiology, Faculty of Veterinary Science, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", University of Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
6
|
Gonzalez-Castro RA, Amoroso-Sanches F, Stokes JE, Graham JK, Carnevale EM. Localisation of phospholipase Cζ1 (PLCZ1) and postacrosomal WW-binding protein (WBP2 N-terminal like) on equine spermatozoa and flow cytometry quantification of PLCZ1 and association with cleavage in vitro. Reprod Fertil Dev 2020; 31:1778-1792. [PMID: 31597592 DOI: 10.1071/rd19217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/03/2019] [Indexed: 02/01/2023] Open
Abstract
Oocyte activation is initiated when a fertilising spermatozoon delivers sperm-borne oocyte-activating factor(s) into the oocyte cytoplasm. Candidates for oocyte activation include two proteins, phospholipase Cζ1 (PLCZ1) and postacrosomal WW-binding protein (PAWP; also known as WBP2 N-terminal like (WBP2NL)). We localised PLCZ1 and WBP2NL/PAWP in stallion spermatozoa and investigated the PLCZ1 content and sperm parameters as well as cleavage after intracytoplasmic sperm injection (ICSI). PLCZ1 was identified as 71-kDa protein in the acrosomal and postacrosomal regions, midpiece and principal piece of the tail. Anti-WBP2NL antibody identified two WBP2NL bands (~28 and ~32kDa) in the postacrosomal region, midpiece and principal piece of the tail. PLCZ1 and WBP2NL expression was positively correlated (P=0.04) in sperm heads. Flow cytometry evaluation of PLCZ1 revealed large variations in fluorescence intensity and the percentage of positively labelled spermatozoa among stallions. PLCZ1 expression was significantly higher in viable than non-viable spermatozoa, and DNA fragmentation was negatively correlated with PLCZ1 expression and the percentage of positively labelled spermatozoa (P<0.05). The use of equine sperm populations considered to have high versus low PLCZ1 content resulted in significantly higher cleavage rates after ICSI of bovine and equine oocytes, supporting the importance of PLCZ1 for oocyte activation.
Collapse
Affiliation(s)
- Raul A Gonzalez-Castro
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, 3101 Rampart Rd, Fort Collins, Colorado, 80521, USA
| | - Fabio Amoroso-Sanches
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, 3101 Rampart Rd, Fort Collins, Colorado, 80521, USA
| | - JoAnne E Stokes
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, 3101 Rampart Rd, Fort Collins, Colorado, 80521, USA
| | - James K Graham
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, 3101 Rampart Rd, Fort Collins, Colorado, 80521, USA
| | - Elaine M Carnevale
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, 3101 Rampart Rd, Fort Collins, Colorado, 80521, USA; and Corresponding author.
| |
Collapse
|
7
|
Kashir J, Mistry BV, BuSaleh L, Abu-Dawas R, Nomikos M, Ajlan A, Abu-Dawud R, AlYacoub N, AlHassan S, Lai FA, Assiri AM, Coskun S. Phospholipase C zeta profiles are indicative of optimal sperm parameters and fertilisation success in patients undergoing fertility treatment. Andrology 2020; 8:1143-1159. [PMID: 32298520 DOI: 10.1111/andr.12796] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/07/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Oocyte activation is driven by intracellular calcium (Ca2+ ) oscillations induced by sperm-specific PLCζ, abrogation of which causes oocyte activation deficiency in humans. Clinical PLCζ investigations have been limited to severe male infertility conditions, while PLCζ levels and localisation patterns have yet to be associated with general sperm viability. MATERIALS AND METHODS PLCζ profiles were examined within a general population of males attending a fertility clinic (65 patients; aged 29-53), examining PLCζ throughout various fractions of sperm viability. Male recruitment criteria required a minimum sperm count of 5 × 106 spermatozoa/mL, while all female patients included in this study yielded at least five oocytes for treatment. Sperm count, motility and semen volume were recorded according to standard WHO reference guidelines and correlated with PLCζ profiles examined via immunoblotting and immunofluorescence. Appropriate fertility treatments were performed following routine clinical standard operating protocols, and fertilisation success determined by successful observation of second polar body extrusion. RESULTS AND DISCUSSION Four distinct PLCζ patterns were observed at the equatorial, acrosomal + equatorial regions of the sperm head, alongside a dispersed pattern, and a population of spermatozoa without any PLCζ. Acrosomal + equatorial PLCζ correlated most to sperm health, while dispersed PLCζ correlated to decreased sperm viability. Total levels of PLCζ exhibited significant correlations with sperm parameters. PLCζ variance corresponded to reduced sperm health, potentially underlying cases of male sub-fertility and increasing male age. Finally, significantly higher levels of PLCζ were exhibited by cases of fertilisation success, alongside higher proportions of Ac + Eq, and lower levels of dispersed PLCζ. CONCLUSIONS PLCζ potentially represents a biomarker of sperm health, and fertilisation capacity in general cases of patients seeking fertility treatment, and not just cases of repeated fertilisation. Further focused investigations are required with larger cohorts to examine the full clinical potential of PLCζ.
Collapse
Affiliation(s)
- Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.,School of Biosciences, Cardiff University, Cardiff, UK.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Bhavesh V Mistry
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Lujain BuSaleh
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Reema Abu-Dawas
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ahmed Ajlan
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Raed Abu-Dawud
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Nadya AlYacoub
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Saad AlHassan
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - F Anthony Lai
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical Research Centre, Qatar University, Doha, Qatar
| | - Abdullah M Assiri
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia.,Biomedical Research Centre, Qatar University, Doha, Qatar.,Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Serdar Coskun
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
8
|
Kashir J. Increasing associations between defects in phospholipase C zeta and conditions of male infertility: not just ICSI failure? J Assist Reprod Genet 2020; 37:1273-1293. [PMID: 32285298 PMCID: PMC7311621 DOI: 10.1007/s10815-020-01748-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Oocyte activation is a fundamental event at mammalian fertilization. In mammals, this process is initiated by a series of characteristic calcium (Ca2+) oscillations, induced by a sperm-specific phospholipase C (PLC) termed PLCzeta (PLCζ). Dysfunction/reduction/deletion of PLCζ is associated with forms of male infertility where the sperm is unable to initiate Ca2+ oscillations and oocyte activation, specifically in cases of fertilization failure. This review article aims to systematically summarize recent advancements and controversies in the field to update expanding clinical associations between PLCζ and various male factor conditions. This article also discusses how such associations may potentially underlie defective embryogenesis and recurrent implantation failure following fertility treatments, alongside potential diagnostic and therapeutic PLCζ approaches, aiming to direct future research efforts to utilize such knowledge clinically. METHODS An extensive literature search was performed using literature databases (PubMed/MEDLINE/Web of Knowledge) focusing on phospholipase C zeta (PLCzeta; PLCζ), oocyte activation, and calcium oscillations, as well as specific male factor conditions. RESULTS AND DISCUSSION Defective PLCζ or PLCζ-induced Ca2+ release can be linked to multiple forms of male infertility including abnormal sperm parameters and morphology, sperm DNA fragmentation and oxidation, and abnormal embryogenesis/pregnancies. Such sperm exhibit absent/reduced levels, and abnormal localization patterns of PLCζ within the sperm head. CONCLUSIONS Defective PLCζ and abnormal patterns of Ca2+ release are increasingly suspected a significant causative factor underlying abnormalities or insufficiencies in Ca2+ oscillation-driven early embryogenic events. Such cases could potentially strongly benefit from relevant therapeutic and diagnostic applications of PLCζ, or even alternative mechanisms, following further focused research efforts.
Collapse
Affiliation(s)
- Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia. .,School of Biosciences, Cardiff University, Cardiff, UK. .,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
9
|
Saleh A, Kashir J, Thanassoulas A, Safieh-Garabedian B, Lai FA, Nomikos M. Essential Role of Sperm-Specific PLC-Zeta in Egg Activation and Male Factor Infertility: An Update. Front Cell Dev Biol 2020; 8:28. [PMID: 32064262 PMCID: PMC7000359 DOI: 10.3389/fcell.2020.00028] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/14/2020] [Indexed: 12/16/2022] Open
Abstract
Sperm-specific phospholipase C zeta (PLCζ) is widely considered to be the physiological stimulus responsible for generating calcium (Ca2+) oscillations that induce egg activation and early embryonic development during mammalian fertilization. In the mammalian testis, PLCζ expression is detected at spermiogenesis following elongated spermatid differentiation. Sperm-delivered PLCζ induces Ca2+ release via the inositol 1,4,5-trisphosphate (InsP3) signaling pathway. PLCζ is the smallest known mammalian PLC isoform identified to date, with the simplest domain organization. However, the distinctive biochemical properties of PLCζ compared with other PLC isoforms contribute to its unique potency in stimulating cytosolic Ca2+ oscillations within mammalian eggs. Moreover, studies describing PLCζ “knockout” mouse phenotypes confirm the supreme importance of PLCζ at egg activation and monospermic fertilization in mice. Importantly, a number of clinical reports have highlighted the crucial importance of PLCζ in human fertilization by associating PLCζ deficiencies with certain forms of male factor infertility. Herein, we give an update on recent advances that have refined our understanding of how sperm PLCζ triggers Ca2 + oscillations and egg activation in mammals, while also discussing the nature of a potential “alternative” sperm factor. We summarise PLCζ localization in mammalian sperm, and the direct links observed between defective PLCζ protein in sperm and documented cases of male infertility. Finally, we postulate how this sperm protein can be used as a potential diagnostic marker, and also as a powerful therapeutic agent for treatment of certain types of male infertility due to egg activation failure or even in more general cases of male subfertility.
Collapse
Affiliation(s)
- Alaaeldin Saleh
- Member of QU Health, College of Medicine, Qatar University, Doha, Qatar
| | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | | | | | - F Anthony Lai
- Member of QU Health, College of Medicine, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| | - Michail Nomikos
- Member of QU Health, College of Medicine, Qatar University, Doha, Qatar
| |
Collapse
|
10
|
Nakamura N, Sloper DT, Del Valle PL. Gene expression profiling of cultured mouse testis fragments treated with ethinylestradiol. J Toxicol Sci 2019; 44:667-679. [PMID: 31588058 DOI: 10.2131/jts.44.667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The assessment of xenobiotic-induced testicular toxicity is important in drug development. Nonetheless, in vitro models to test drugs and chemicals that may cause testicular toxicity are lacking, requiring the continued use of animal models for those studies. We previously evaluated an in vitro mouse testis organ culture system using ethinylestradiol (EE), a well-studied testicular toxicant, and demonstrated a dose-dependent relationship between adverse effects to germ cell differentiation and increasing EE concentrations. However, we terminated that study after 20 days of culture due to oxygen deficiency during germ cell differentiation. Therefore, in the current study, we aimed to identify gene(s) with potential for supporting the histopathological evaluations of testicular toxicity using in vitro testis organ culture system. We cultured testis fragments obtained from mice at postnatal day (PND) 5 in α-Minimal Essential Medium containing 40 mg/mL AlbuMAX™ I and treated them with 0.01 or 1 nM EE on day 1 of culture. On day 20, we collected testis fragments for RNA sequencing analysis and quantitative polymerase chain reaction (qPCR). We found that phospholipase C, zeta 1 and testis-specific serine kinase 4 genes, that are involved in spermatogenesis and predominantly expressed in the testis, were significantly reduced in testis fragments treated with the highest concentration of EE. Also, cytochrome P450, family 26, subfamily b, polypeptide 1 (Cyp26b1) and interleukin 16 (Il16) were up-regulated in the highest EE-treated groups. Further studies are needed to confirm the variations of these gene expression using other testicular toxicants.
Collapse
Affiliation(s)
- Noriko Nakamura
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration, USA
| | - Daniel T Sloper
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration, USA
| | - Pedro L Del Valle
- Center for Drug Evaluation and Research, Food and Drug Administration, USA
| |
Collapse
|
11
|
Kashir J, Nomikos M, Lai FA. Phospholipase C zeta and calcium oscillations at fertilisation: The evidence, applications, and further questions. Adv Biol Regul 2017; 67:148-162. [PMID: 29108881 DOI: 10.1016/j.jbior.2017.10.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 12/26/2022]
Abstract
Oocyte activation is a fundamental event at mammalian fertilisation, initiated by a series of characteristic calcium (Ca2+) oscillations in mammals. This characteristic pattern of Ca2+ release is induced in a species-specific manner by a sperm-specific enzyme termed phospholipase C zeta (PLCζ). Reduction or absence of functional PLCζ within sperm underlies male factor infertility in humans, due to mutational inactivation or abrogation of PLCζ protein expression. Underlying such clinical implications, a significant body of evidence has now been accumulated that has characterised the unique biochemical and biophysical properties of this enzyme, further aiding the unique clinical opportunities presented. Herein, we present and discuss evidence accrued over the past decade and a half that serves to support the identity of PLCζ as the mammalian sperm factor. Furthermore, we also discuss the potential novel avenues that have yet to be examined regarding PLCζ mechanism of action in both the oocyte, and the sperm. Finally, we discuss the advances that have been made regarding the clinical therapeutic and diagnostic applications of PLCζ in potentially treating male infertility as a result of oocyte activation deficiency (OAD), and also possibly more general cases of male subfertility.
Collapse
Affiliation(s)
- Junaid Kashir
- College of Biomedical & Life Sciences, School of Biosciences, Cardiff University, Cardiff, UK; Alfaisal University, College of Medicine, Riyadh, Saudi Arabia; King Faisal Specialist Hospital & Research Center, Department of Comparative Medicine, Riyadh, Saudi Arabia.
| | - Michail Nomikos
- College of Medicine, Member of QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - F Anthony Lai
- College of Biomedical & Life Sciences, School of Biosciences, Cardiff University, Cardiff, UK; College of Medicine, Member of QU Health, Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
12
|
The role and mechanism of action of sperm PLC-zeta in mammalian fertilisation. Biochem J 2017; 474:3659-3673. [PMID: 29061915 DOI: 10.1042/bcj20160521] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 12/26/2022]
Abstract
At mammalian fertilisation, the fundamental stimulus that triggers oocyte (egg) activation and initiation of early embryonic development is an acute rise of the intracellular-free calcium (Ca2+) concentration inside the egg cytoplasm. This essential Ca2+ increase comprises a characteristic series of repetitive Ca2+ oscillations, starting soon after sperm-egg fusion. Over the last 15 years, accumulating scientific and clinical evidence supports the notion that the physiological stimulus that precedes the cytosolic Ca2+ oscillations is a novel, testis-specific phospholipase C (PLC) isoform, known as PLC-zeta (PLCζ). Sperm PLCζ catalyses the hydrolysis of phosphatidylinositol 4,5-bisphosphate triggering cytosolic Ca2+ oscillations through the inositol 1,4,5-trisphosphate signalling pathway. PLCζ is the smallest known mammalian PLC isoform with the most elementary domain organisation. However, relative to somatic PLCs, the PLCζ isoform possesses a unique potency in stimulating Ca2+ oscillations in eggs that is attributed to its novel biochemical characteristics. In this review, we discuss the latest developments that have begun to unravel the vital role of PLCζ at mammalian fertilisation and decipher its unique mechanism of action within the fertilising egg. We also postulate the significant potential diagnostic and therapeutic capacity of PLCζ in alleviating certain types of male infertility.
Collapse
|
13
|
Azad N, Nazarian H, Ghaffari Novin M, Masteri Farahani R, Piryaei A, Heidari MH. Phospholipase C zeta parameters in sperm from polymorphic teratozoospermic men. Ann Anat 2017; 215:63-70. [PMID: 28954206 DOI: 10.1016/j.aanat.2017.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 09/01/2017] [Accepted: 09/08/2017] [Indexed: 01/16/2023]
Abstract
Teratozoospermia is a disorder associated with high abnormal sperm morphology which affects fertility in males. In recent years, it has been described that biomarker-based sperm quality evaluation can alleviate male infertility treatment. Phospholipase C zeta (PLCζ) is a sperm-specific factor which appears to be a predicting biomarker for fertilization potential of males. Following fertilization, PLCζ enters into oocyte cytoplasm and induces oocyte activation, a fundamental stage in initiation of embryo development. Currently, PLCζ parameters, including localization patterns, the proportion of PLCζ-expressing sperm and the expression level, are not defined in polymorphic teratozoospermic men. This study aimed to evaluate PLCζ parameters in polymorphic teratozoospermic men, and compare these parameters with fertile normozoospermic men. Semen samples from thirteen normozoospermic fertile men and twenty-three polymorphic teratozoospermic men were included in this study and evaluated using western blotting and immunofluorescence analyses. Our data indicated significantly lower expression of PLCζ in polymorphic teratozoospermic men, as compared with control men; however, there was no significant difference in localization patterns and the proportion of PLCζ-expressing sperm between polymorphic teratozoospermic patients and control men. Collectively, findings from the present study demonstrated that polymorphic teratozoospermic men did not show abnormal localization patterns or the absence of PLCζ, as compared to the control men; nonetheless, lower expression of PLCζ, considering its role in oocyte activation, might be one of the possible causes of infertility in these patients.
Collapse
Affiliation(s)
- Nahid Azad
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Nazarian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; IVF Center, Taleghani Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marefat Ghaffari Novin
- Infertility and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Masteri Farahani
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Heidari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Hachem A, Godwin J, Ruas M, Lee HC, Ferrer Buitrago M, Ardestani G, Bassett A, Fox S, Navarrete F, de Sutter P, Heindryckx B, Fissore R, Parrington J. PLCζ is the physiological trigger of the Ca 2+ oscillations that induce embryogenesis in mammals but conception can occur in its absence. Development 2017; 144:2914-2924. [PMID: 28694258 DOI: 10.1242/dev.150227] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/01/2017] [Indexed: 12/21/2022]
Abstract
Activation of the egg by the sperm is the first, vital stage of embryogenesis. The sperm protein PLCζ has been proposed as the physiological agent that triggers the Ca2+ oscillations that normally initiate embryogenesis. Consistent with this, recombinant PLCζ induces Ca2+ oscillations in eggs and debilitating mutations in the PLCZ1 gene are associated with infertility in men. However, there has been no evidence that knockout of the gene encoding PLCζ abolishes the ability of sperm to induce Ca2+ oscillations in eggs. Here, we show that sperm derived from Plcz1-/- male mice fail to trigger Ca2+ oscillations in eggs, cause polyspermy and thus demonstrate that PLCζ is the physiological trigger of these Ca2+ oscillations. Remarkably, some eggs fertilized by PLCζ-null sperm can develop, albeit at greatly reduced efficiency, and after a significant time-delay. In addition, Plcz1-/- males are subfertile but not sterile, suggesting that in the absence of PLCζ, spontaneous egg activation can eventually occur via an alternative route. This is the first demonstration that in vivo fertilization without the normal physiological trigger of egg activation can result in offspring. PLCζ-null sperm now make it possible to resolve long-standing questions in fertilization biology, and to test the efficacy and safety of procedures used to treat human infertility.
Collapse
Affiliation(s)
- Alaa Hachem
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Jonathan Godwin
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Margarida Ruas
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Hoi Chang Lee
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - Minerva Ferrer Buitrago
- Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Goli Ardestani
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - Andrew Bassett
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sebastian Fox
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Felipe Navarrete
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - Petra de Sutter
- Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Björn Heindryckx
- Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Rafael Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
15
|
Ray PF, Toure A, Metzler-Guillemain C, Mitchell MJ, Arnoult C, Coutton C. Genetic abnormalities leading to qualitative defects of sperm morphology or function. Clin Genet 2016; 91:217-232. [PMID: 27779748 DOI: 10.1111/cge.12905] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 12/12/2022]
Abstract
Infertility, defined by the inability of conceiving a child after 1 year is estimated to concern approximately 50 million couples worldwide. As the male gamete is readily accessible and can be studied by a simple spermogram it is easier to subcategorize male than female infertility. Subjects with a specific sperm phenotype are more likely to have a common origin thus facilitating the search for causal factors. Male infertility is believed to be often multifactorial and caused by both genetic and extrinsic factors, but severe cases of male infertility are likely to have a predominant genetic etiology. Patients presenting with a monomorphic teratozoospermia such as globozoospermia or macrospermia with more than 85% of the spermatozoa presenting this specific abnormality have been analyzed permitting to identify several key genes for spermatogenesis such as AURKC and DPY19L2. The study of patients with other specific sperm anomalies such as severe alteration of sperm motility, in particular multiple morphological anomalies of the sperm flagella (MMAF) or sperm unability to fertilize the oocyte (oocyte activation failure syndrome) has also enable the identification of new infertility genes. Here we review the recent works describing the identification and characterization of gene defects having a direct qualitative effect on sperm morphology or function.
Collapse
Affiliation(s)
- P F Ray
- Université Grenoble Alpes, Grenoble, France.,Institut for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Grenoble, France.,UF de Biochimie et Génétique Moléculaire, CHU Grenoble Alpes, Grenoble, France
| | - A Toure
- Institut Cochin, INSERM U1016, Paris, France.,CNRS, UMR8104, Paris, France.,Sorbonne Paris Cité, Faculté de Médecine, Université Paris Descartes, Paris, France
| | | | | | - C Arnoult
- Université Grenoble Alpes, Grenoble, France.,Institut for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Grenoble, France
| | - C Coutton
- Université Grenoble Alpes, Grenoble, France.,Institut for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Grenoble, France.,UF de Génétique Chromosomique, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
16
|
Tavalaee M, Kiani-Esfahani A, Nasr-Esfahani MH. Relationship between Potential Sperm Factors Involved in Oocyte Activation and Sperm DNA Fragmentation with Intra-Cytoplasmic Sperm Injection Clinical Outcomes. CELL JOURNAL 2016; 18:588-596. [PMID: 28042543 PMCID: PMC5086337 DOI: 10.22074/cellj.2016.4725] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/31/2016] [Indexed: 12/30/2022]
Abstract
Objective The present study aimed to simultaneously evaluate the association between expression of three potential factors [post-acrosomal sheath WW domain-binding protein (PAWP), phospholipase Cζ (PLCζ), and truncated form of the kit receptor
(TR-KIT)] as candidates of oocyte activation with fertilization rate and early embryonic development.
Materials and Methods In this experimental study, semen samples were collected
from 35 intra-cytoplasmic sperm injection (ICSI) candidates and analyzed according to World Health Organization criteria (2010). Each sample was divided into two
parts. The first part was processed for insemination by density-gradient centrifugation (DGC) and the second part was prepared for assessment of sperm morphology (Papanicolaou staining), DNA fragmentation [transferase dUTP nick end labeling
(TUNEL)], and three Sperm-borne oocyte-activating factor (s) (SOAFs)-PLCζ, PAWP,
and TR-KIT.
Results Significant positive correlations existed between the percentages of PLCζ,
PAWP, and TR-KIT with fertilization rate. In addition, significant negative correlations
existed between the percentage of DNA fragmentation with the percentages of PLCζ
and PAWP. We did not find a relationship between percentages of PLCζ, PAWP, and
TR-KIT with embryo quality and pregnancy rate (P>0.05). There was a significant
negative correlation between percentage of DNA fragmentation with fertilization and
embryo quality.
Conclusion Oocyte activation was associated with the studied sperm factors (PAWP,
PLCζ, and TR-KIT). These factors might hold the potential to be considered as diagnostic
factors in the assessment of semen samples to evaluate their potential to induce oocyte
activation. In addition, we observed a significant association between DNA fragmentation
with fertilization, as well as embryo quality and expression of PAWP and PLCζ, which
indicated that men with high degrees of DNA fragmentation might require artificial oocyte
activation. Whether such action should take place, and its cost and benefits should be
evaluated in the future.
Collapse
Affiliation(s)
- Marziyeh Tavalaee
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Abbas Kiani-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Isfahan Fertility and Infertility Center, Isfahan, Iran
| |
Collapse
|
17
|
Kamali-Dolat Abadi M, Tavalaee M, Shahverdi A, Nasr-Esfahani MH. Evaluation of PLCζ and PAWP Expression in Globozoospermic Individuals. CELL JOURNAL 2016; 18:438-45. [PMID: 27602326 PMCID: PMC5011332 DOI: 10.22074/cellj.2016.4572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 01/17/2016] [Indexed: 11/28/2022]
Abstract
Objective Globozoospermia is a rare type of teratozoospermia with incidence of 0.1%
among infertile individuals. Phospholipase C zeta (PLCζ) and postacrosomal sheath WW
domain binding protein (PAWP) are the main candidates in sperm taking responsibility for
oocyte activation during fertilization. Therefore, we aimed to evaluate the expression of
these two genes at RNA and protein levels in globozoospermic individuals and compare
the results with fertile individuals.
Materials and Methods In this experimental study, semen samples of 21 infertile
men with globozoospermia and 25 fertile men were collected. Expression of PLCζ
and PAWP at RNA and protein levels were assessed and compared between two
groups by quantitative real time polymerase chain reaction (qPCR) and Western blot,
respectively.
Results Expression of both PLCζ and PAWP were significantly reduced at RNA and protein levels in infertile men with globozoospermia compared to fertile men.
Conclusion This is the first study that simultaneously assessing the respective factors in
a large population of globozoospermia, suggested that intra-cytoplasmic sperm injection
(ICSI) along with artificial oocyte activation may rescue failed fertilization in routine ICSI.
Collapse
Affiliation(s)
- Majid Kamali-Dolat Abadi
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Department of Molecular and Cellular Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Marziyeh Tavalaee
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Isfahan Fertility and Infertility Center, Isfahan, Iran
| |
Collapse
|
18
|
Sanders JR, Swann K. Molecular triggers of egg activation at fertilization in mammals. Reproduction 2016; 152:R41-50. [PMID: 27165049 DOI: 10.1530/rep-16-0123] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/09/2016] [Indexed: 01/15/2023]
Abstract
In mammals, the sperm activates the development of the egg by triggering a series of oscillations in the cytosolic-free Ca(2+) concentration (Ca(2+) i). The sperm triggers these cytosolic Ca(2+i) oscillations after sperm-egg membrane fusion, as well as after intracytoplasmic sperm injection (ICSI). These Ca(2+) i oscillations are triggered by a protein located inside the sperm. The identity of the sperm protein has been debated over many years, but all the repeatable data now suggest that it is phospholipase Czeta (PLCζ). The main downstream target of Ca(2+) i oscillations is calmodulin-dependent protein kinase II (CAMKII (CAMK2A)), which phosphorylates EMI2 and WEE1B to inactivate the M-phase promoting factor protein kinase activity (MPF) and this ultimately triggers meiotic resumption. A later decline in the activity of mitogen-activated protein kinase (MAPK) then leads to the completion of activation which is marked by the formation of pronuclei and entry into interphase of the first cell cycle. The early cytosolic Ca(2+) increases also trigger exocytosis via a mechanism that does not involve CAMKII. We discuss some recent developments in our understanding of these triggers for egg activation within the framework of cytosolic Ca(2+) signaling.
Collapse
Affiliation(s)
| | - Karl Swann
- School of BiosciencesCardiff University, Cardiff, UK
| |
Collapse
|
19
|
Escoffier J, Lee HC, Yassine S, Zouari R, Martinez G, Karaouzène T, Coutton C, Kherraf ZE, Halouani L, Triki C, Nef S, Thierry-Mieg N, Savinov SN, Fissore R, Ray PF, Arnoult C. Homozygous mutation of PLCZ1 leads to defective human oocyte activation and infertility that is not rescued by the WW-binding protein PAWP. Hum Mol Genet 2016; 25:878-91. [PMID: 26721930 PMCID: PMC4754041 DOI: 10.1093/hmg/ddv617] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/06/2015] [Accepted: 12/17/2015] [Indexed: 11/13/2022] Open
Abstract
In mammals, sperm-oocyte fusion initiates Ca(2+) oscillations leading to a series of events called oocyte activation, which is the first stage of embryo development. Ca(2+) signaling is elicited by the delivery of an oocyte-activating factor by the sperm. A sperm-specific phospholipase C (PLCZ1) has emerged as the likely candidate to induce oocyte activation. Recently, PAWP, a sperm-born tryptophan domain-binding protein coded by WBP2NL, was proposed to serve the same purpose. Here, we studied two infertile brothers exhibiting normal sperm morphology but complete fertilization failure after intracytoplasmic sperm injection. Whole exomic sequencing evidenced a missense homozygous mutation in PLCZ1, c.1465A>T; p.Ile489Phe, converting Ile 489 into Phe. We showed the mutation is deleterious, leading to the absence of the protein in sperm, mislocalization of the protein when injected in mouse GV and MII oocytes, highly abnormal Ca(2+) transients and early embryonic arrest. Altogether these alterations are consistent with our patients' sperm inability to induce oocyte activation and initiate embryo development. In contrast, no deleterious variants were identified in WBP2NL and PAWP presented normal expression and localization. Overall we demonstrate in humans, the absence of PLCZ1 alone is sufficient to prevent oocyte activation irrespective of the presence of PAWP. Additionally, it is the first mutation located in the C2 domain of PLCZ1, a domain involved in targeting proteins to cell membranes. This opens the door to structure-function studies to identify the conserved amino acids of the C2 domain that regulate the targeting of PLCZ1 and its selectivity for its lipid substrate(s).
Collapse
Affiliation(s)
- Jessica Escoffier
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | | | - Sandra Yassine
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Raoudha Zouari
- Polyclinique les Jasmins, Centre d'Aide Médicale à la Procréation, Centre Urbain Nord, 1003 Tunis, Tunisia
| | - Guillaume Martinez
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Thomas Karaouzène
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Charles Coutton
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, CHU de Grenoble, UF de Génétique Chromosomique, Grenoble F-38000, France
| | - Zine-Eddine Kherraf
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France
| | - Lazhar Halouani
- Polyclinique les Jasmins, Centre d'Aide Médicale à la Procréation, Centre Urbain Nord, 1003 Tunis, Tunisia
| | - Chema Triki
- Clinique Hannibal, Centre d'AMP, les berges du lac, 1053 Tunis, Tunisia
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Nicolas Thierry-Mieg
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, Laboratoire TIMC-IMAG, UMR CNRS 5525, Grenoble F-38000, France and
| | - Sergey N Savinov
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | | | - Pierre F Ray
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France, CHU de Grenoble, UF de Biochimie et Génétique Moléculaire, Grenoble F-38000, France
| | - Christophe Arnoult
- Université Grenoble Alpes, Grenoble, F-38000, Grenoble, France, Institut Albert Bonniot, INSERM U823, La Tronche F-38700, France,
| |
Collapse
|
20
|
Javadian-Elyaderani S, Ghaedi K, Tavalaee M, Rabiee F, Deemeh MR, Nasr-Esfahani MH. Diagnosis of genetic defects through parallel assessment of PLCζ and CAPZA3 in infertile men with history of failed oocyte activation. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:281-9. [PMID: 27114798 PMCID: PMC4834118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 12/24/2015] [Indexed: 11/12/2022]
Abstract
OBJECTIVES Phospholipase C ζ (PLCζ) is considered as a nominee for sperm associated oocyte activating factors and is located back-to-back with CAPZA3, an actin-capping protein controlling actin polymerization during spermiogenesis. They contain a common bidirectional promoter. The objective of this study was to identify individuals with parallel low expression of PLCζ and CAPZA3 mRNA, in hope of detecting genetic defects in this bidirectional promoter. MATERIALS AND METHODS Semen samples were collected from 24 fertile and 59 infertile individuals with total failed, low and high fertilization rate post intra-cytoplasmic sperm injection (ICSI), as well as globozoospermic individuals. Expression of PLCζ and CAPZA3 were assessed by Real time PCR. In addition, PLCζ was assessed by Western blot. RESULTS Significant correlations between PLCζ with CAPZA3 and also between these two genes with fertilization were observed. Individuals with low fertilization presented significantly lower expression of these two genes. Low expression of PLCζ was also verified by Western analysis. Sequence analysis of bidirectional promoter of these two genes in an individual with parallel low expression of both PLCζ and CAPZA3, revealed a mutation within the CAPZA3 predicted promoter, known as human regulatory factor X4 which is a testis-specific dimeric DNA-binding protein. In the opposite stand, in the same location, the mutation appears to be outside but in the vicinity of PLCζ, in a binding region predicate by Genomatix. CONCLUSION Parallel assessment of CAPZA3 with PLCζ at mRNA level in individuals with inability to induce oocyte activation may help researcher to identify genetic defects associated with failed fertilization.
Collapse
Affiliation(s)
- Soudabeh Javadian-Elyaderani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kamran Ghaedi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Biology Department, School of Sciences, University of Isfahan, Isfahan, Iran
| | - Marziyeh Tavalaee
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Farzaneh Rabiee
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Reza Deemeh
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Isfahan Fertility and Infertility Center, Isfahan, Iran
| |
Collapse
|
21
|
Anifandis G, Messini CI, Dafopoulos K, Daponte A, Messinis IE. Sperm contributions to oocyte activation: more that meets the eye. J Assist Reprod Genet 2016; 33:313-316. [PMID: 26780328 DOI: 10.1007/s10815-016-0653-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/04/2016] [Indexed: 11/24/2022] Open
Abstract
It is well known that for successful fertilization, oocyte activation is required, which involves a signal transduction cascade leading to the conversion of the oocyte to a diploid embryo. During oocyte activation, intracellular calcium levels oscillate repetitively causing exocytosis of cortical granules, the enzymes which the latter contain are released into the perivitelline space, leading to modifications of the zona pellucida (ZP), which prevent the penetration of the ZP by further spermatozoa. The necessary element that initiates oocyte activation is apparently the release of intracellular calcium (Ca(2+)) stored in the endoplasmic reticulum (ER). The exact mechanism via which Ca(2+) is released within the oocyte has not been yet clarified, and has been a matter of an ongoing debate. Today, the sperm factor hypothesis has gained general acceptance, according to which a sperm molecule, either phospholipase C (PLCζ) or a post-acrosomal sheath WW domain-binding protein (PAWP), diffuses into the ooplasm initiating a molecular cascade involving mainly the phosphoinositide pathway. Mounting evidence now indicates that these calcium oscillations are caused by a testis-specific PLC termed PLCζ, released into the oocyte following gamete fusion. Also, recently, PAWP has been proposed as an alternative sperm factor candidate. These different sperm candidates have led to a significant debate. This raises important questions as regards to the relative importance of these two proteins as diagnostic tools in reproductive medicine with therapeutic potential, indicating the need for further research. In the present mini review, the phenomenon of oocyte activation during fertilization as well as the existing controversy will be highlighted and the possible mechanisms that are involved in this process will be discussed. Finally, an explanation of the existing debate will be attempted.
Collapse
Affiliation(s)
- George Anifandis
- Department of Obstetrics and Gynaecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larissa, Greece.
| | - Christina I Messini
- Department of Obstetrics and Gynaecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larissa, Greece
| | - Konstantinos Dafopoulos
- Department of Obstetrics and Gynaecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larissa, Greece
| | - Alexandros Daponte
- Department of Obstetrics and Gynaecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larissa, Greece
| | - Ioannis E Messinis
- Department of Obstetrics and Gynaecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larissa, Greece
| |
Collapse
|
22
|
Abstract
The most fundamental unresolved issue of fertilization is to define how the sperm activates the egg to begin embryo development. Egg activation at fertilization in all species thus far examined is caused by some form of transient increase in the cytoplasmic free Ca2+ concentration. What has not been clear, however, is precisely how the sperm triggers the large changes in Ca2+ observed within the egg cytoplasm. Here, we review the studies indicating that the fertilizing sperm stimulates a cytosolic Ca2+ increase in the egg specifically by delivering a soluble factor that diffuses into the cytosolic space of the egg upon gamete membrane fusion. Evidence is primarily considered in species of eggs where the sperm has been shown to elicit a cytosolic Ca2+ increase by initiating Ca2+ release from intracellular Ca2+ stores. We suggest that our best understanding of these signaling events is in mammals, where the sperm triggers a prolonged series of intracellular Ca2+ oscillations. The strongest empirical studies to date suggest that mammalian sperm-triggered Ca2+ oscillations are caused by the introduction of a sperm-specific protein, called phospholipase C-zeta (PLCζ) that generates inositol trisphosphate within the egg. We will discuss the role and mechanism of action of PLCζ in detail at a molecular and cellular level. We will also consider some of the evidence that a soluble sperm protein might be involved in egg activation in nonmammalian species.
Collapse
Affiliation(s)
- Karl Swann
- College of Biomedical and Life Sciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - F. Anthony Lai
- College of Biomedical and Life Sciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
23
|
Amdani SN, Yeste M, Jones C, Coward K. Phospholipase C zeta (PLCζ) and male infertility: Clinical update and topical developments. Adv Biol Regul 2015; 61:58-67. [PMID: 26700242 DOI: 10.1016/j.jbior.2015.11.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 01/09/2023]
Abstract
The development of a mammalian embryo is initiated by a sequence of molecular events collectively referred to as 'oocyte activation' and regulated by the release of intracellular calcium in the ooplasm. Over the last decade, phospholipase C zeta (PLCζ), a sperm protein introduced into the oocyte upon gamete fusion, has gained almost universal acceptance as the protein factor responsible for initiating oocyte activation. A large body of consistent and reproducible evidence, from both biochemical and clinical settings, confers support for the role of PLCζ in this fundamental biological context, which has significant ramifications for the management of human male infertility. Oocyte activation deficiency (OAD) and total fertilisation failure (TFF) are known causes of infertility and have both been linked to abnormalities in the structure, expression, and localisation pattern of PLCζ in human sperm. Assisted oocyte activators (AOAs) represent the only therapeutic option available for OAD at present, although these agents have been the source of much debate recently, particularly with regard to their potential epigenetic effects upon the embryo. Consequently, there is much interest in the deployment of sensitive PLCζ assays as prognostic/diagnostic tests and human recombinant PLCζ protein as an alternative form of therapy. Although PLCζ deficiency has been directly linked to a cohort of infertile cases, we have yet to identify the specific causal mechanisms involved. While two genetic mutations have been identified which link defective PLCζ protein to an infertile phenotype, both were observed in the same patient, and have yet to be described in other patients. Consequently, some researchers are investigating the possibility that genetic variations in the form of single nucleotide polymorphisms (SNPs) could provide some explanation, especially since >6000 SNPs have been identified in the PLCζ gene. As yet, however, there is no consistent data to suggest that any of these SNPs influence the functional ability of PLCζ. Other laboratories appear to be focussing upon the PLCζ promoter, which is bi-directional and shared with the actin filament capping muscle Z-line alpha 3 gene (CAPZA3), or seeking to identify interacting proteins within the ooplasm. The aim of this review is to provide a synopsis of recent progress in the application of PLCζ in diagnostic and therapeutic medicine, to discuss our current understanding of how the functional ability of PLCζ might be controlled, and thus how PLCζ deficiency might arise, and finally, to consider the potential implications of alternative sperm protein candidates, such as post-acrosomal WW-domain binding protein (PAWP), which has caused much debate and confusion in the field over the last few years.
Collapse
Affiliation(s)
- Siti Nornadhirah Amdani
- Nuffield Department of Obstetrics & Gynaecology, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK; PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tunku Link, Gadong, Brunei Darussalam
| | - Marc Yeste
- Nuffield Department of Obstetrics & Gynaecology, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Celine Jones
- Nuffield Department of Obstetrics & Gynaecology, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Kevin Coward
- Nuffield Department of Obstetrics & Gynaecology, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK.
| |
Collapse
|
24
|
Yeste M, Jones C, Amdani SN, Patel S, Coward K. Oocyte activation deficiency: a role for an oocyte contribution? Hum Reprod Update 2015; 22:23-47. [DOI: 10.1093/humupd/dmv040] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/13/2015] [Indexed: 12/11/2022] Open
|
25
|
Amdani SN, Yeste M, Jones C, Coward K. Sperm Factors and Oocyte Activation: Current Controversies and Considerations1. Biol Reprod 2015; 93:50. [DOI: 10.1095/biolreprod.115.130609] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/30/2015] [Indexed: 11/01/2022] Open
|
26
|
Abstract
Mammalian embryo development is init iated by intracel lular Ca2+ oscillations that result in oocyte activation following gamete membrane fusion. It is widely believed that oocyte Ca2+ oscillations are triggered by a sperm-specific protein, phospholipase C-zeta (PLCζ) that activates InsP3 production leading to repetitive Ca2+ release from intracellular stores. However, a recent report in the FASEB Journal by Aarabi et al. challenges this view by proposing postacrosomal WW domain-binding protein (PAWP) as another sperm-derived protein that can also initiate Ca2+ oscillations and zygotic development at fertilization. Here we discuss these new findings and examine the evidence suggesting PAWP as the "real" sperm factor.
Collapse
Affiliation(s)
| | | | - Richard Oko
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, ON, Canada,
| |
Collapse
|
27
|
Aarabi M, Balakier H, Bashar S, Moskovtsev SI, Sutovsky P, Librach CL, Oko R. Sperm‐derived WW domain‐binding protein, PAWP, elicits calcium oscillations and oocyte activation in humans and mice. FASEB J 2014; 28:4434-40. [DOI: 10.1096/fj.14-256495] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Mahmoud Aarabi
- Department of Biomedical and Molecular SciencesSchool of MedicineQueen's UniversityKingstonOntarioCanada
- Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| | | | | | - Sergey I. Moskovtsev
- CReATe Fertility CentreTorontoOntarioCanada
- Department of Obstetrics and GynecologyUniversity of TorontoTorontoOntarioCanada
| | - Peter Sutovsky
- Division of Animal SciencesGynecology and Women's HealthSchool of MedicineUniversity of MissouriColumbiaMissouriUSA
- Department of ObstetricsGynecology and Women's HealthSchool of MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Clifford L. Librach
- CReATe Fertility CentreTorontoOntarioCanada
- Department of Obstetrics and GynecologyUniversity of TorontoTorontoOntarioCanada
| | - Richard Oko
- Department of Biomedical and Molecular SciencesSchool of MedicineQueen's UniversityKingstonOntarioCanada
| |
Collapse
|
28
|
Aarabi M, Balakier H, Bashar S, Moskovtsev SI, Sutovsky P, Librach CL, Oko R. Sperm content of postacrosomal WW binding protein is related to fertilization outcomes in patients undergoing assisted reproductive technology. Fertil Steril 2014; 102:440-7. [PMID: 24907910 DOI: 10.1016/j.fertnstert.2014.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 05/01/2014] [Accepted: 05/01/2014] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To determine the levels of postacrosomal WW binding protein (PAWP) in the spermatozoa of men that were used clinically for intracytoplasmic sperm injection (ICSI) and to correlate them with infertility treatment outcomes. DESIGN Prospective clinical and laboratory study. SETTING University-based laboratory and infertility clinic. PATIENT(S) Men undergoing ICSI for the treatment of couples' infertility (n=110). INTERVENTION(S) Quantitative analysis of sperm PAWP levels by flow cytometry and developmental analysis of PAWP expression by immunoblotting, immunofluorescence, and immunohistochemistry. MAIN OUTCOME MEASURE(S) PAWP flow-cytometric levels and immunolocalization in spermatozoa. RESULT(S) A strong positive correlation was found between PAWP expression levels and fertilization rates after ICSI, with high levels of PAWP being associated with higher fertilization rates; the positive correlation was independent of age, DNA fragmentation index, and other sperm parameters. PAWP expression levels were correlated with embryonic development, with high levels of PAWP being associated with a lower number of arrested embryos within 3-5 days post-ICSI. PAWP expression was detected during the late stages of human spermiogenesis in elongating spermatids, confirming previous findings in various animal models. CONCLUSION(S) Our clinical data from infertile couples demonstrate significant correlations between sperm PAWP levels and both fertilization rates and normal embryonic development after ICSI. Considering its proposed role in the initiation of oocyte activation, we suggest that PAWP could have potential applications in the diagnosis and treatment of infertility.
Collapse
Affiliation(s)
- Mahmoud Aarabi
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | | | - Sergey I Moskovtsev
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, Missouri; Departments of Obstetrics, Gynecology, and Women's Health, School of Medicine, University of Missouri, Columbia, Missouri
| | - Clifford L Librach
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| | - Richard Oko
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
29
|
Kashir J, Nomikos M, Lai FA, Swann K. Sperm-induced Ca2+ release during egg activation in mammals. Biochem Biophys Res Commun 2014; 450:1204-11. [PMID: 24769204 DOI: 10.1016/j.bbrc.2014.04.078] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/15/2014] [Indexed: 12/12/2022]
Abstract
This review discusses the role that the sperm-specific phospholipase C zeta (PLCζ) is proposed to play during the fertilization of mammalian eggs. At fertilization, the sperm initiates development by causing a series of oscillations in cytosolic concentrations of calcium [Ca(2)] within the egg. PLCζ mimics the sperm at fertilization, causing the same pattern of Ca(2+) release as seen at fertilization. Introducing PLCζ into mouse eggs also mimics a number of other features of the way in which the fertilizing sperm triggers Ca(2+) oscillations. We discuss the localization of PLCζ within the egg and present a hypothesis about the localization of PLCζ within the sperm before the initiation of fertilization.
Collapse
Affiliation(s)
- Junaid Kashir
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Michail Nomikos
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - F Anthony Lai
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Karl Swann
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| |
Collapse
|
30
|
Vanden Meerschaut F, Nikiforaki D, Heindryckx B, De Sutter P. Assisted oocyte activation following ICSI fertilization failure. Reprod Biomed Online 2014; 28:560-71. [PMID: 24656559 DOI: 10.1016/j.rbmo.2014.01.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 01/12/2014] [Accepted: 01/22/2014] [Indexed: 10/25/2022]
Abstract
The capacity of intracytoplasmic sperm injection (ICSI) to permit almost any type of spermatozoa to fertilize oocytes has made it the most successful treatment for male factor infertility. Despite its high success rates, fertilization failure following ICSI still occurs in 1-3% of couples. Assisted oocyte activation (AOA) is being increasingly applied in human assisted reproduction to restore fertilization and pregnancy rates in couples with a history of ICSI fertilization failure. However, controversy still exists mainly because the artificial activating agents do not mimic precisely the initial physiological processes of mammalian oocyte activation, which has led to safety concerns. This review addresses the mechanism of human oocyte activation and the relatively rare phenomenon of fertilization failure after ICSI. Next, it describes the current diagnostic approaches and focuses on the application, efficiency and safety of AOA in human assisted reproduction.
Collapse
Affiliation(s)
- Frauke Vanden Meerschaut
- Department for Reproductive Medicine, University Hospital Ghent, De Pintelaan 185 - 1P4, 9000 Ghent, Belgium
| | - Dimitra Nikiforaki
- Department for Reproductive Medicine, University Hospital Ghent, De Pintelaan 185 - 1P4, 9000 Ghent, Belgium
| | - Björn Heindryckx
- Department for Reproductive Medicine, University Hospital Ghent, De Pintelaan 185 - 1P4, 9000 Ghent, Belgium.
| | - Petra De Sutter
- Department for Reproductive Medicine, University Hospital Ghent, De Pintelaan 185 - 1P4, 9000 Ghent, Belgium
| |
Collapse
|
31
|
Neri QV, Lee B, Rosenwaks Z, Machaca K, Palermo GD. Understanding fertilization through intracytoplasmic sperm injection (ICSI). Cell Calcium 2013; 55:24-37. [PMID: 24290744 DOI: 10.1016/j.ceca.2013.10.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 10/28/2013] [Accepted: 10/31/2013] [Indexed: 01/21/2023]
Abstract
Since the establishment of in vitro fertilization, it became evident that almost half of the couples failed to achieve fertilization and this phenomenon was attributed to a male gamete dysfunction. The adoption of assisted fertilization techniques particularly ICSI has been able to alleviate male factor infertility by granting the consistent ability of a viable spermatozoon to activate an oocyte. Single sperm injection, by pinpointing the beginning of fertilization, has been an invaluable tool in clarifying the different aspects of early fertilization and syngamy. However, even with ICSI some couples fail to fertilize due to ooplasmic dysmaturity in relation to the achieved nuclear maturation marked by the extrusion of the first polar body. More uncommon are cases where the spermatozoa partially or completely lack the specific oocyte activating factor. In this work, we review the most relevant aspects of fertilization and its failure through assisted reproductive technologies. Attempts at diagnosing and treating clinical fertilization failure are described.
Collapse
Affiliation(s)
- Queenie V Neri
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Bora Lee
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Zev Rosenwaks
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Education City - Qatar Foundation, Doha, Qatar
| | - Gianpiero D Palermo
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
32
|
Nomikos M, Kashir J, Swann K, Lai FA. Sperm PLCζ: from structure to Ca2+ oscillations, egg activation and therapeutic potential. FEBS Lett 2013; 587:3609-16. [PMID: 24157362 DOI: 10.1016/j.febslet.2013.10.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/12/2013] [Accepted: 10/14/2013] [Indexed: 10/26/2022]
Abstract
Significant evidence now supports the assertion that cytosolic calcium oscillations during fertilization in mammalian eggs are mediated by a testis-specific phospholipase C (PLC), termed PLC-zeta (PLCζ) that is released into the egg following gamete fusion. Herein, we describe the current paradigm of PLCζ in this fundamental biological process, summarizing recent important advances in our knowledge of the biochemical and physiological properties of this enzyme. We describe the data suggesting that PLCζ has distinct features amongst PLCs enabling the hydrolysis of its substrate, phosphatidylinositol 4,5-bisphosphate (PIP2) at low Ca(2+) levels. PLCζ appears to be unique in its ability to target PIP2 that is present on intracellular vesicles. We also discuss evidence that PLCζ may be a significant factor in human fertility with potential therapeutic capacity.
Collapse
Affiliation(s)
- Michail Nomikos
- Institute of Molecular and Experimental Medicine, WHRI, Cardiff University School of Medicine, Heath Park, CF144XN Cardiff, UK.
| | | | | | | |
Collapse
|
33
|
Amdani SN, Jones C, Coward K. Phospholipase C zeta (PLCζ): oocyte activation and clinical links to male factor infertility. Adv Biol Regul 2013; 53:292-308. [PMID: 23916605 DOI: 10.1016/j.jbior.2013.07.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 06/02/2023]
Abstract
Mounting scientific and clinical evidence supports the key role played by phospholipase C zeta (PLCζ), a sperm-specific protein, in the activation of oocytes following fertilisation. Lacking a pleckstrin homology domain, PLCζ remains the smallest known mammalian PLC and was first identified in 2002. Since then, PLCζ has been the target for a multitude of studies in both mammalian and non-mammalian species focused upon its fundamental biochemical activity and crucial role as the mediator of oocyte activation. The earliest event subsequent to gamete fusion is the onset of a series of intracellular calcium oscillations within the oocyte, which are known to modulate cortical granule exocytosis, release meiotic arrest, regulate gene expression, recruit maternal mRNA, and initiate embryogenesis. Collectively these processes are known as 'oocyte activation' and together, represent a fundamental mechanism for early embryonic development. Evidence suggests that these processes are initiated and controlled by calcium release from ooplasmic sources in response to PLCζ activity via the inositol-1,4,5-triphosphate (IP3) pathway. While the biochemical action of PLCζ has been extensively studied, especially in relation to the EF-hands, X-Y linker, and C2 domain, all of which play critical roles for in vivo activity, there are still key gaps in our knowledge, particularly in terms of regulation and interaction with other proteins within the oocyte. Moreover, increasing clinical evidence has revealed a strong correlation between certain types of male infertility and the aberrant expression, localisation, structure and function of PLCζ in human sperm, particularly in cases of recurrent intracytoplasmic sperm injection (ICSI) failure, globozoospermia, and oocyte activation deficiency (OAD). In addition, two heterozygous substitution mutations have been identified in the coding sequence of PLCζ in one particular patient causing disruption to the catalytic X and Y domains and resulting in infertility. Although, such cases can be treated via the use of artificial oocyte activators (AOAs) such as calcium ionophores, significant concern remains over the use of such chemical agents, largely due to the fact that calcium release manifests as a single transient, rather than a series of oscillations as observed during normal fertilisation. Current interest in PLCζ is thus to develop a series of prognostic, diagnostic and therapeutic approaches which could first identify male patients that are deficient in PLCζ and then rescue oocyte activation ability via assisted reproductive technology (ART) and a pure, functionally-active, recombinant human PLCζ protein. While significant progress has been made in such areas over recent years, there is a clear need to translate scientific findings to clinical settings in order to maximise successful outcome for patients.
Collapse
Affiliation(s)
- Siti Nornadhirah Amdani
- Nuffield Department of Obstetrics & Gynaecology, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | | | | |
Collapse
|
34
|
Kashir J, Deguchi R, Jones C, Coward K, Stricker SA. Comparative biology of sperm factors and fertilization-induced calcium signals across the animal kingdom. Mol Reprod Dev 2013; 80:787-815. [PMID: 23900730 DOI: 10.1002/mrd.22222] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/23/2013] [Indexed: 11/08/2022]
Abstract
Fertilization causes mature oocytes or eggs to increase their concentrations of intracellular calcium ions (Ca²⁺) in all animals that have been examined, and such Ca²⁺ elevations, in turn, provide key activating signals that are required for non-parthenogenetic development. Several lines of evidence indicate that the Ca²⁺ transients produced during fertilization in mammals and other taxa are triggered by soluble factors that sperm deliver into oocytes after gamete fusion. Thus, for a broad-based analysis of Ca²⁺ dynamics during fertilization in animals, this article begins by summarizing data on soluble sperm factors in non-mammalian species, and subsequently reviews various topics related to a sperm-specific phospholipase C, called PLCζ, which is believed to be the predominant activator of mammalian oocytes. After characterizing initiation processes that involve sperm factors or alternative triggering mechanisms, the spatiotemporal patterns of Ca²⁺ signals in fertilized oocytes or eggs are compared in a taxon-by-taxon manner, and broadly classified as either a single major transient or a series of repetitive oscillations. Both solitary and oscillatory types of fertilization-induced Ca²⁺ signals are typically propagated as global waves that depend on Ca²⁺ release from the endoplasmic reticulum in response to increased concentrations of inositol 1,4,5-trisphosphate (IP₃). Thus, for taxa where relevant data are available, upstream pathways that elevate intraoocytic IP3 levels during fertilization are described, while other less-common modes of producing Ca²⁺ transients are also examined. In addition, the importance of fertilization-induced Ca²⁺ signals for activating development is underscored by noting some major downstream effects of these signals in various animals.
Collapse
Affiliation(s)
- Junaid Kashir
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford, UK
| | | | | | | | | |
Collapse
|
35
|
Vanden Meerschaut F, D'Haeseleer E, Gysels H, Thienpont Y, Dewitte G, Heindryckx B, Oostra A, Roeyers H, Van Lierde K, De Sutter P. Neonatal and neurodevelopmental outcome of children aged 3-10 years born following assisted oocyte activation. Reprod Biomed Online 2013; 28:54-63. [PMID: 24125944 DOI: 10.1016/j.rbmo.2013.07.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 07/18/2013] [Accepted: 07/23/2013] [Indexed: 12/20/2022]
Abstract
Assisted oocyte activation (AOA) using a calcium ionophore has been used for more than a decade following intracytoplasmic sperm injection (ICSI) fertilization failure. However, since AOA does not mimic precisely the physiological fertilization process, concerns exist about its use in human assisted reproduction. This study assessed the neonatal and neurodevelopmental outcome of children aged ≥ 3 years who had been born following AOA in our centre. Twenty-one children participated in the study (81% response rate; mean age 63.6 ± 21.07 months). Neonatal data were collected via questionnaires. Neurodevelopmental outcome was tested using the Reynell Developmental Language Scales or Clinical Evaluation of Language Fundamentals, Wechsler Preschool and Primary Scale of Intelligence or Wechsler Intelligence Scale for Children, and the Movement Assessment Battery for Children III. Behaviour was scored by the Social Communication Questionnaire, the Child Behaviour Checklist and the Teachers Report Form. For all tests and questionnaires, the mean outcomes lay within the expected ranges. These are first data on the developmental outcome of AOA children. The high response rate and the robustness of the tests support the data, which are reassuring although still considered preliminary. Therefore, AOA should still be performed only in selected couples.
Collapse
Affiliation(s)
- Frauke Vanden Meerschaut
- Department for Reproductive Medicine, University Hospital Ghent, De Pintelaan 185-1P4, 9000 Ghent, Belgium.
| | - Evelien D'Haeseleer
- Department of Otorhinolaryngology and Head and Neck Surgery, Speech and Language Pathology, University Hospital Ghent, De Pintelaan 185-2P2, 9000 Ghent, Belgium
| | - Hannelore Gysels
- Department of Experimental Clinical and Health Psychology, Research Group Developmental Disorders, Ghent University, Henri Dunantlaan 2, 9000 Ghent, Belgium
| | - Ylenia Thienpont
- Department of Experimental Clinical and Health Psychology, Research Group Developmental Disorders, Ghent University, Henri Dunantlaan 2, 9000 Ghent, Belgium
| | - Griet Dewitte
- Centre for Developmental Disorders, De Pintelaan 185-2K5, 9000 Ghent, Belgium
| | - Björn Heindryckx
- Department for Reproductive Medicine, University Hospital Ghent, De Pintelaan 185-1P4, 9000 Ghent, Belgium
| | - An Oostra
- Centre for Developmental Disorders, De Pintelaan 185-2K5, 9000 Ghent, Belgium
| | - Herbert Roeyers
- Department of Experimental Clinical and Health Psychology, Research Group Developmental Disorders, Ghent University, Henri Dunantlaan 2, 9000 Ghent, Belgium
| | - Kristiane Van Lierde
- Department of Otorhinolaryngology and Head and Neck Surgery, Speech and Language Pathology, University Hospital Ghent, De Pintelaan 185-2P2, 9000 Ghent, Belgium
| | - Petra De Sutter
- Department for Reproductive Medicine, University Hospital Ghent, De Pintelaan 185-1P4, 9000 Ghent, Belgium
| |
Collapse
|
36
|
Vanden Meerschaut F, Nikiforaki D, De Roo C, Lierman S, Qian C, Schmitt-John T, De Sutter P, Heindryckx B. Comparison of pre- and post-implantation development following the application of three artificial activating stimuli in a mouse model with round-headed sperm cells deficient for oocyte activation. Hum Reprod 2013; 28:1190-8. [DOI: 10.1093/humrep/det038] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
37
|
Sobinoff AP, Sutherland JM, Mclaughlin EA. Intracellular signalling during female gametogenesis. Mol Hum Reprod 2012; 19:265-78. [PMID: 23247812 DOI: 10.1093/molehr/gas065] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Female reproductive potential is dictated by the size of the primordial follicle pool and the correct regulation of oocyte maturation and activation--events essential for production of viable offspring. Although a substantial body of work underpins our understanding of these processes, the molecular mechanisms of follicular and oocyte development are not fully understood. This review summarizes recent findings which have improved our conception of how folliculogenesis and oocyte competence are regulated, and discusses their implications for assisted reproductive techniques. We highlight evidence provided by genetically modified mouse models and in vitro studies which have refined our understanding of Pi3k/Akt and mTOR signalling in the oocyte and have discovered a role for Jak/Stat/Socs signalling in granulosa cells during primordial follicle activation. We also appraise a novel role for the metal ion zinc in the regulation of meiosis I and meiosis II progression through early meiosis inhibitor (Emi2) and Mos-Mapk signalling, and examine studies which expand our understanding of intracellular calcium signalling and extrinsic Plcζ in stimulating oocyte activation.
Collapse
Affiliation(s)
- A P Sobinoff
- Priority Research Centre in Chemical Biology, School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW2308, Australia
| | | | | |
Collapse
|
38
|
Ramadan WM, Kashir J, Jones C, Coward K. Oocyte activation and phospholipase C zeta (PLCζ): diagnostic and therapeutic implications for assisted reproductive technology. Cell Commun Signal 2012; 10:12. [PMID: 22591604 PMCID: PMC3393615 DOI: 10.1186/1478-811x-10-12] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/16/2012] [Indexed: 12/18/2022] Open
Abstract
Infertility affects one in seven couples globally and has recently been classified as a disease by the World Health Organisation (WHO). While in-vitro fertilisation (IVF) offers effective treatment for many infertile couples, cases exhibiting severe male infertility (19-57%) often remain difficult, if not impossible to treat. In such cases, intracytoplasmic sperm injection (ICSI), a technique in which a single sperm is microinjected into the oocyte, is implemented. However, 1-5% of ICSI cycles still fail to fertilise, affecting over 1000 couples per year in the UK alone. Pregnancy and delivery rates for IVF and ICSI rarely exceed 30% and 23% respectively. It is therefore imperative that Assisted Reproductive Technology (ART) protocols are constantly modified by associated research programmes, in order to provide patients with the best chances of conception. Prior to fertilisation, mature oocytes are arrested in the metaphase stage of the second meiotic division (MII), which must be alleviated to allow the cell cycle, and subsequent embryogenesis, to proceed. Alleviation occurs through a series of concurrent events, collectively termed 'oocyte activation'. In mammals, oocytes are activated by a series of intracellular calcium (Ca2+) oscillations following gamete fusion. Recent evidence implicates a sperm-specific phospholipase C, PLCzeta (PLCζ), introduced into the oocyte following membrane fusion as the factor responsible. This review summarises our current understanding of oocyte activation failure in human males, and describes recent advances in our knowledge linking certain cases of male infertility with defects in PLCζ expression and activity. Systematic literature searches were performed using PubMed and the ISI-Web of Knowledge. Databases compiled by the United Nations and World Health Organisation databases (UNWHO), and the Human Fertilization and Embryology Authority (HFEA) were also scrutinised. It is clear that PLCζ plays a fundamental role in the activation of mammalian oocytes, and that genetic, molecular, or biochemical perturbation of this key enzyme is strongly linked to human infertility where oocyte activation is deficient. Consequently, there is significant scope for our understanding of PLCζ to be translated to the ART clinic, both as a novel therapeutic agent with which to rescue oocyte activation deficiency (OAD), or as a prognostic/diagnostic biomarker of oocyte activation ability in target sperm samples.
Collapse
Affiliation(s)
- Walaa M Ramadan
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Headington, Oxford, OX3, 9DU, UK
| | - Junaid Kashir
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Headington, Oxford, OX3, 9DU, UK
| | - Celine Jones
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Headington, Oxford, OX3, 9DU, UK
| | - Kevin Coward
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Headington, Oxford, OX3, 9DU, UK
| |
Collapse
|