1
|
Ioannou D, Tempest HG. The genetic basis of male and female infertility. Syst Biol Reprod Med 2025; 71:143-169. [PMID: 40294233 DOI: 10.1080/19396368.2025.2493621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025]
Abstract
This review provides a comprehensive overview of the genetic factors underlying male and female infertility. Infertility affects an estimated one in six couples worldwide, with both male and female factors contributing equally to its prevalence. Approximately, 50% of infertility cases are attributed to genetic causes. We explore three main categories of genetic causes: chromosomal abnormalities, monogenic disorders, and syndromic conditions. Chromosomal causes, including numerical and structural aberrations, are discussed with a focus on their impact on gametogenesis and reproductive outcomes. We review key monogenic causes of infertility, highlighting recent discoveries in genes critical for gonadal development, gametogenesis, and hormonal regulation. Syndromic conditions affecting fertility are examined, highlighting their impact on reproductive function. Throughout the review, we address the challenges in identifying genetic mechanisms of infertility, particularly focusing on the intricate processes involved in oogenesis and spermatogenesis. We also discuss how advancements in genetic testing, such as next-generation sequencing (NGS) and genome-wide association studies (GWAS), have significantly enhanced our understanding of idiopathic infertility and promise further insights in the future. We also discuss the clinical implications of genetic diagnoses, including the role of preimplantation genetic testing (PGT) and genetic counseling in reproductive medicine. This review synthesizes current knowledge on the genetic basis of infertility, providing a comprehensive overview of chromosomal, monogenic, and syndromic causes. It aims to offer readers a solid foundation for understanding the complex genetic factors underlying reproductive disorders.
Collapse
Affiliation(s)
- Dimitrios Ioannou
- Department of Basic Sciences, College of Medicine, Roseman University of Health Sciences, Las Vegas, NV, USA
| | - Helen G Tempest
- Department of Basic Sciences, College of Medicine, Roseman University of Health Sciences, Las Vegas, NV, USA
| |
Collapse
|
2
|
Al-Ali H, Baig A, Alkhanjari RR, Murtaza ZF, Alhajeri MM, Elbahrawi R, Abdukadir A, Bhamidimarri PM, Kashir J, Hamdan H. Septins as key players in spermatogenesis, fertilisation and pre-implantation embryogenic cytoplasmic dynamics. Cell Commun Signal 2024; 22:523. [PMID: 39468561 PMCID: PMC11514797 DOI: 10.1186/s12964-024-01889-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Septins are a family of cytokinesis-related proteins involved in regulating cytoskeletal design, cell morphology, and tissue morphogenesis. Apart from cytokinesis, as a fourth component of cytoskeleton, septins aid in forming scaffolds, vesicle sorting and membrane stability. They are also known to be involved in the regulation of intracellular calcium (Ca2+) via the STIM/Orai complex. Infertility affects ~ 15% of couples globally, while male infertility affects ~ 7% of men. Global pregnancy and live birth rates following fertility treatment remain relatively low, while there has been an observable decline in male fertility parameters over the past 60 years. Low fertility treatment success can be attributed to poor embryonic development, poor sperm parameters and fertilisation defects. While studies from the past few years have provided evidence for the role of septins in fertility related processes, the functional role of septins and its related complexes in cellular processes such as oocyte activation, fertilization, and sperm maturation are not completely understood. This review summarizes the available knowledge on the role of septins in spermatogenesis and oocyte activation via Ca2+ regulation, and cytoskeletal dynamics throughout pre-implantation embryonic development. We aim to identify the currently less known mechanisms by which septins regulate these immensely important mechanisms with a view of identifying areas of investigation that would benefit our understanding of cell and reproductive biology, but also provide potential avenues to improve current methods of fertility treatment.
Collapse
Affiliation(s)
- Hana Al-Ali
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Amna Baig
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Rayyah R Alkhanjari
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Zoha F Murtaza
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Maitha M Alhajeri
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Rawdah Elbahrawi
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Azhar Abdukadir
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Poorna Manasa Bhamidimarri
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Junaid Kashir
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates.
- Center for Biotechnology, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
| | - Hamdan Hamdan
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates.
| |
Collapse
|
3
|
Mehdinejadiani S, Goudarzi N, Masjedi F, Govahi A, Mirani M, Mehdinejadiani K, Azad N. Clinical Applications of Assisted Oocyte Activation in Couples with Various Reproductive Problems: A Systematic Review. Reprod Sci 2024; 31:2916-2942. [PMID: 39168919 DOI: 10.1007/s43032-024-01671-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024]
Abstract
ICSI may face fertilization failure, prompting the use of assisted oocyte activation (AOA) techniques. While AOA is implemented in infertility clinics, its target patients and definitive application remain uncertain. This systematic review adheres to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines to assess reproductive outcomes in ICSI-AOA cycles compared to conventional ICSI and evaluate AOA effectiveness in various infertility disorders. A literature search encompassed PubMed, Web of Science, EMBASE, and Scopus databases until December 2023 for relevant English studies. Included studies compared ICSI-AOA with conventional ICSI in couples with prior fertilization failure, utilizing diverse AOA methods. Control groups consisted of sibling oocytes, previous cycles of the same couples, or couples undergoing conventional ICSI. Evaluated outcomes included fertilization, cleavage, embryo quality, implantation, pregnancy, and live birth rates. Article screening and data extraction were performed by two authors, with risk of bias assessed by another investigator. Out of 3088 initially identified articles, 30 studies were included, focusing on fertilization failure (n = 10), female infertility (n = 3), PLCζ defects (n = 4), poor sperm quality (n = 4), Globozoospermia (n = 4), and surgically retrieved sperm (n = 8). Most studies concluded that AOA could overcome fertilization failure, but success rates varied based on sperm-related or oocyte-related factors in ICSI-AOA cycles. Due to differences in patient inclusion criteria and sample sizes, most studies were not sufficiently similar for pooled analysis, limiting robust conclusions. There is insufficient evidence, particularly from randomized controlled trials (RCTs), to determine the efficacy or safety of ICSI-AOA as a treatment strategy. Registration number is PROSPERO, CRD42024551221.
Collapse
Affiliation(s)
- Shayesteh Mehdinejadiani
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasim Goudarzi
- Department of Anatomical Sciences, Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Fatemeh Masjedi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azam Govahi
- Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Mirani
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kobra Mehdinejadiani
- Department of Microbiology, Immunology and Infectious Diseases, Calgary Univercity, Calgary, Canada
| | - Nahid Azad
- Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Mostafa Khomeini Blvd, Semnan, Iran.
| |
Collapse
|
4
|
Kashir J, Mistry BV, Rajab MA, BuSaleh L, Abu-Dawud R, Ahmed HA, Alharbi S, Nomikos M, AlHassan S, Coskun S, Assiri AM. The mammalian sperm factor phospholipase C zeta is critical for early embryo division and pregnancy in humans and mice. Hum Reprod 2024; 39:1256-1274. [PMID: 38670547 PMCID: PMC11145019 DOI: 10.1093/humrep/deae078] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
STUDY QUESTION Are sperm phospholipase C zeta (PLCζ) profiles linked to the quality of embryogenesis and pregnancy? SUMMARY ANSWER Sperm PLCζ levels in both mouse and humans correlate with measures of ideal embryogenesis whereby minimal levels seem to be required to result in successful pregnancy. WHAT IS KNOWN ALREADY While causative factors underlying male infertility are multivariable, cases are increasingly associated with the efficacy of oocyte activation, which in mammals occurs in response to specific profiles of calcium (Ca2+) oscillations driven by sperm-specific PLCζ. Although sperm PLCζ abrogation is extensively linked with human male infertility where oocyte activation is deficient, less is clear as to whether sperm PLCζ levels or localization underlies cases of defective embryogenesis and failed pregnancy following fertility treatment. STUDY DESIGN, SIZE, DURATION A cohort of 54 couples undergoing fertility treatment were recruited at the assisted reproductive technology laboratory at the King Faisal Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia. The recruitment criteria for males was a minimum sperm concentration of 5×106 sperm/ml, while all female patients had to have at least five oocytes. Sperm PLCζ analysis was performed in research laboratories, while semen assessments were performed, and time-lapse morphokinetic data were obtained, in the fertility clinic as part of routine treatment. The CRISPR/Cas9 system was concurrently used to induce indels and single-nucleotide mutations within the Plcζ gene to generate strains of Plcζ mutant mice. Sperm PLCζ was evaluated using immunofluorescence and immunoblotting with an antibody of confirmed consistent specificity against PLCζ. PARTICIPANTS/MATERIALS, SETTING, METHODS We evaluated PLCζ profiles in sperm samples from 54 human couples undergoing fertility treatment in the context of time-lapse morphokinetic analysis of resultant embryos, correlating such profiles to pregnancy status. Concurrently, we generated two strains of mutant Plcζ mice using CRISPR/Cas9, and performed IVF with wild type (WT) oocytes and using WT or mutant Plcζ sperm to generate embryos. We also assessed PLCζ status in WT and mutant mice sperm in the context of time-lapse morphokinetic analysis and breeding outcomes. MAIN RESULTS AND THE ROLE OF CHANCE A significant (P ≤ 0.05) positive relationship was observed between both PLCζ relative fluorescence and relative density with the times taken for both the second cell division (CC2) (r = 0.26 and r = 0.43, respectively) and the third cell division (S2) (r = 0.26). Examination of localization patterns also indicated significant correlations between the presence or absence of sperm PLCζ and CC2 (r = 0.27 and r = -0.27, respectively; P ≤ 0.025). Human sperm PLCζ levels were at their highest in the ideal times of CC2 (8-12 h) compared to time ranges outside the ideal timeframe (<8 and >12 h) where levels of human sperm PLCζ were lower. Following assignment of PLCζ level thresholds, quantification revealed a significantly higher (P ≤ 0.05) rate of successful pregnancy in values larger than the assigned cut-off for both relative fluorescence (19% vs 40%, respectively) and relative density (8% vs 54%, respectively). Immunoblotting indicated a single band for PLCζ at 74 kDa in sperm from WT mice, while a single band was also observed in sperm from heterozygous of Plcζ mutant mouse sperm, but at a diminished intensity. Immunofluorescent analysis indicated the previously reported (Kashir et al., 2021) fluorescence patterns in WT sperm, while sperm from Plcζ mutant mice exhibited a significantly diminished and dispersed pattern at the acrosomal region of the sperm head. Breeding experiments indicated a significantly reduced litter size of mutant Plcζ male mice compared to WT mice, while IVF-generated embryos using sperm from mutant Plcζ mice exhibited high rates of polyspermy, and resulted in significantly reduced numbers of these embryos reaching developmental milestones. LIMITATIONS, REASONS FOR CAUTION The human population examined was relatively small, and should be expanded to examine a larger multi-centre cohort. Infertility conditions are often multivariable, and it was not possible to evaluate all these in human patients. However, our mutant Plcζ mouse experiments do suggest that PLCζ plays a significant role in early embryo development. WIDER IMPLICATIONS OF THE FINDINGS We found that minimal levels of PLCζ within a specific range were required for optimal early embryogenesis, correlating with increased pregnancy. Levels of sperm PLCζ below specific thresholds were associated with ineffective embryogenesis and lower pregnancy rates, despite eliciting successful fertilization in both mice and humans. To our knowledge, this represents the first time that PLCζ levels in sperm have been correlated to prognostic measures of embryogenic efficacy and pregnancy rates in humans. Our data suggest for the first time that the clinical utilization of PLCζ may stand to benefit not just a specific population of male infertility where oocyte activation is completely deficient (wherein PLCζ is completely defective/abrogated), but also perhaps the larger population of couples seeking fertility treatment. STUDY FUNDING/COMPETING INTEREST(S) J.K. is supported by a faculty start up grant awarded by Khalifa University (FSU-2023-015). This study was also supported by a Healthcare Research Fellowship Award (HF-14-16) from Health and Care Research Wales (HCRW) to J.K., alongside a National Science, Technology, and Innovation plan (NSTIP) project grant (15-MED4186-20) awarded by the King Abdulaziz City for Science and Technology (KACST) for J.K. and A.M.A. The authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Junaid Kashir
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Bhavesh V Mistry
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohamed A Rajab
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Lujain BuSaleh
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Raed Abu-Dawud
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Institute for Molecular Medicine, MSH Medical School, Hamburg, Germany
| | - Hala A Ahmed
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sarah Alharbi
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Saad AlHassan
- Department of Obstetrics and Gynaecology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Serdar Coskun
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdullah M Assiri
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Sheibak N, Amjadi F, Shamloo A, Zarei F, Zandieh Z. Microfluidic sperm sorting selects a subpopulation of high-quality sperm with a higher potential for fertilization. Hum Reprod 2024; 39:902-911. [PMID: 38461455 DOI: 10.1093/humrep/deae045] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/12/2024] [Indexed: 03/12/2024] Open
Abstract
STUDY QUESTION Is a microfluidic sperm sorter (MSS) able to select higher quality sperm compared to conventional methods? SUMMARY ANSWER The MSS selects sperm with improved parameters, lower DNA fragmentation, and higher fertilizing potential. WHAT IS KNOWN ALREADY To date, the few studies that have compared microfluidics sperm selection with conventional methods have used heterogeneous study population and have lacked molecular investigations. STUDY DESIGN, SIZE, DURATION The efficiency of a newly designed MSS in isolating high-quality sperm was compared to the density-gradient centrifugation (DGC) and swim-up (SU) methods, using 100 semen samples in two groups, during 2023-2024. PARTICIPANTS/MATERIALS, SETTING, METHODS Semen specimens from 50 normozoospermic and 50 non-normozoospermic men were sorted using MSS, DGC, and SU methods to compare parameters related to the quality and fertilizing potential of sperm. The fertilizing potential of sperm was determined by measurement of phospholipase C zeta (PLCζ) and post-acrosomal sheath WW domain-binding protein (PAWP) expression using flow cytometry, and the chromatin dispersion test was used to assess sperm DNA damage. MAIN RESULTS AND THE ROLE OF CHANCE In both normozoospermic and non-normozoospermic groups, the MSS-selected sperm with the highest progressive motility, PLCζ positive expression and PLCζ and PAWP fluorescence intensity the lowest non-progressive motility, and minimal DNA fragmentation, compared to sperm selected by DGC and SU methods (P < 0.05). LIMITATION, REASONS FOR CAUTION The major limitations of our study were the low yield of sperm in the MSS chips and intentional exclusion of severe male factor infertility to yield a sufficient sperm count for molecular experiments; thus testing with severe oligozoospermic semen and samples with low count and motility is still required. In addition, due to ethical considerations, at present, it was impossible to use the sperm achieved from MSS in the clinic to assess the fertilization rate and further outcomes. WIDER IMPLICATIONS OF THE FINDINGS Our research presents new evidence that microfluidic sperm sorting may result in the selection of high-quality sperm from raw semen. This novel technology might be a key to improving clinical outcomes of assisted reproduction in infertile patients. STUDY FUNDING/COMPETING INTEREST(S) The study is funded by the Iran University of Medical Sciences and no competing interest exists. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Nadia Sheibak
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Amjadi
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Amir Shamloo
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Fatemeh Zarei
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran
| | - Zahra Zandieh
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Chou CW, Chen SU, Chang CH, Tsai YY, Huang CC. Aggressive sperm immobilization improves reproductive outcomes in patients with suboptimal semen parameters and previous ICSI fertilization failure. Sci Rep 2024; 14:5363. [PMID: 38438506 PMCID: PMC10912663 DOI: 10.1038/s41598-024-56092-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/01/2024] [Indexed: 03/06/2024] Open
Abstract
Intracytoplasmic sperm injection (ICSI) is the most effective procedure to resolve male infertility, enhancing overall fertilization and pregnancy outcomes. However, it is important to note that fertilization failure (FF) can still occur in a few cases after ICSI. This study aims to introduce a specialized technique of aggressive sperm immobilization for ICSI and evaluate its impact on reproductive outcomes in cases involving prior fertilization failure. All infertile couples with male partners having suboptimal semen samples and previous ICSI fertilization failure were evaluated using retrospective data from National Taiwan tertiary university hospital (NTUH) between January 2016 and February 2022. Fertilization failure in our study was defined as less than 30% fertilization rate (FR, the number of normally fertilized oocytes divided by the total number of injected mature oocytes). Data involving both standard (routine procedure) and aggressive sperm immobilization (SI) techniques during different ICSI cycles were included in this study. Standard and aggressive SI methods were performed by compressing the distal half tail of the spermatozoa ≦ 5 and 15 times prior to ICSI respectively. Generalized estimating equations analysis were applied to compare the clinical outcomes between two procedures. Overall, data from 23 infertile couples who had undergone 65 ICSI cycles (31 standard SI with low fertilization rate and 34 aggressive SI) were included in the study. The average FR in the ICSI cycles with standard SI and aggressive SI were 23.6 ± 23.1% and 49.5 ± 31.8 respectively (P = 0.0002). The majority of embryos were transferred at the day 3 stage, with an average number transferred of 2.6 ± 0.9 in the aggressive SI group and 1.9 ± 0.9 in the standard group. The number of embryos transferred per transfer cycle was higher in the aggressive SI (P = 0.015), whereas the number of good-quality embryos was similar between the two procedures (P = 0.44). There were one and seven live births from the standard SI cycles and aggressive SI cycles respectively. In conclusion, aggressive SI was associated with a significantly higher FR, resulting in more available embryos for transfer without compromising embryo quality. Therefore, this specialized technique improved pregnancy outcome among infertile couples with a previous ICSI-FF. It can be a safe, economic, and effective method to improve the assisted reproductive technologies outcomes for infertile patients affected by previous ICSI-FF.
Collapse
Affiliation(s)
- Ching-Wen Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Shee-Uan Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Chin-Hao Chang
- Department of Medical Research, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Yi Tsai
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Chu-Chun Huang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, 100, Taiwan.
| |
Collapse
|
7
|
Wei Y, Wang J, Qu R, Zhang W, Tan Y, Sha Y, Li L, Yin T. Genetic mechanisms of fertilization failure and early embryonic arrest: a comprehensive review. Hum Reprod Update 2024; 30:48-80. [PMID: 37758324 DOI: 10.1093/humupd/dmad026] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/07/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Infertility and pregnancy loss are longstanding problems. Successful fertilization and high-quality embryos are prerequisites for an ongoing pregnancy. Studies have proven that every stage in the human reproductive process is regulated by multiple genes and any problem, at any step, may lead to fertilization failure (FF) or early embryonic arrest (EEA). Doctors can diagnose the pathogenic factors involved in FF and EEA by using genetic methods. With the progress in the development of new genetic technologies, such as single-cell RNA analysis and whole-exome sequencing, a new approach has opened up for us to directly study human germ cells and reproductive development. These findings will help us to identify the unique mechanism(s) that leads to FF and EEA in order to find potential treatments. OBJECTIVE AND RATIONALE The goal of this review is to compile current genetic knowledge related to FF and EEA, clarifying the mechanisms involved and providing clues for clinical diagnosis and treatment. SEARCH METHODS PubMed was used to search for relevant research articles and reviews, primarily focusing on English-language publications from January 1978 to June 2023. The search terms included fertilization failure, early embryonic arrest, genetic, epigenetic, whole-exome sequencing, DNA methylation, chromosome, non-coding RNA, and other related keywords. Additional studies were identified by searching reference lists. This review primarily focuses on research conducted in humans. However, it also incorporates relevant data from animal models when applicable. The results were presented descriptively, and individual study quality was not assessed. OUTCOMES A total of 233 relevant articles were included in the final review, from 3925 records identified initially. The review provides an overview of genetic factors and mechanisms involved in the human reproductive process. The genetic mutations and other genetic mechanisms of FF and EEA were systematically reviewed, for example, globozoospermia, oocyte activation failure, maternal effect gene mutations, zygotic genome activation abnormalities, chromosome abnormalities, and epigenetic abnormalities. Additionally, the review summarizes progress in treatments for different gene defects, offering new insights for clinical diagnosis and treatment. WIDER IMPLICATIONS The information provided in this review will facilitate the development of more accurate molecular screening tools for diagnosing infertility using genetic markers and networks in human reproductive development. The findings will also help guide clinical practice by identifying appropriate interventions based on specific gene mutations. For example, when an individual has obvious gene mutations related to FF, ICSI is recommended instead of IVF. However, in the case of genetic defects such as phospholipase C zeta1 (PLCZ1), actin-like7A (ACTL7A), actin-like 9 (ACTL9), and IQ motif-containing N (IQCN), ICSI may also fail to fertilize. We can consider artificial oocyte activation technology with ICSI to improve fertilization rate and reduce monetary and time costs. In the future, fertility is expected to be improved or restored by interfering with or supplementing the relevant genes.
Collapse
Affiliation(s)
- Yiqiu Wei
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingxuan Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Qu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiqian Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiling Tan
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanwei Sha
- Department of Andrology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Bafleh WS, Abdulsamad HMR, Al-Qaraghuli SM, El Khatib RY, Elbahrawi RT, Abdukadir AM, Alsawae SM, Dimassi Z, Hamdan H, Kashir J. Applications of advances in mRNA-based platforms as therapeutics and diagnostics in reproductive technologies. Front Cell Dev Biol 2023; 11:1198848. [PMID: 37305677 PMCID: PMC10250609 DOI: 10.3389/fcell.2023.1198848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
The recent COVID-19 pandemic led to many drastic changes in not only society, law, economics, but also in science and medicine, marking for the first time when drug regulatory authorities cleared for use mRNA-based vaccines in the fight against this outbreak. However, while indeed representing a novel application of such technology in the context of vaccination medicine, introducing RNA into cells to produce resultant molecules (proteins, antibodies, etc.) is not a novel principle. It has been common practice to introduce/inject mRNA into oocytes and embryos to inhibit, induce, and identify several factors in a research context, while such aspects have also been proposed as potential therapeutic and diagnostic applications to combat infertility in humans. Herein, we describe key areas where mRNA-based platforms have thus far represented potential areas of clinical applications, describing the advantages and limitations of such applications. Finally, we also discuss how recent advances in mRNA-based platforms, driven by the recent pandemic, may stand to benefit the treatment of infertility in humans. We also present brief future directions as to how we could utilise recent and current advancements to enhance RNA therapeutics within reproductive biology, specifically with relation to oocyte and embryo delivery.
Collapse
Affiliation(s)
- Wjdan S. Bafleh
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Haia M. R. Abdulsamad
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Sally M. Al-Qaraghuli
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Riwa Y. El Khatib
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Rawdah Taha Elbahrawi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Azhar Mohamud Abdukadir
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | | | - Zakia Dimassi
- Department of Pediatrics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Junaid Kashir
- Department of Biology, College of Arts and Science, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Lin Y, Huang Y, Li B, Zhang T, Niu Y, Hu S, Ding Y, Yao G, Wei Z, Yao N, Yao Y, Lu Y, He Y, Zhu Q, Zhang L, Sun Y. Novel mutations in PLCZ1 lead to early embryonic arrest as a male factor. Front Cell Dev Biol 2023; 11:1193248. [PMID: 37261077 PMCID: PMC10227596 DOI: 10.3389/fcell.2023.1193248] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Early embryonic arrest is one of the causes of assist reproduction technology (ART) failure. We have previously reported that the first sperm-derived genetic factor, ACTL7a mutations, could lead to early embryonic arrest. However, whether there are other male genetic factors associated with early embryonic arrest remains elusive. Here, we reported bi-allelic mutations in PLCZ1, a well-known causal gene of total fertilization failure, in four infertile males. Among these mutations, p.403_404del, p.I489S, and p.W536X were newly reported in this study. Histological and Western blotting analysis of the patients' sperm indicated these variants as loss-of-function mutations. These patients manifested normal conventional semen parameters and ultra-structures in sperm heads. However, among four in vitro fertilization (IVF) cycles, 81.8% (18/22) of the oocytes were polyspermic fertilized, which was rarely reported in PLCZ1-related male patients. In the following six ICSI cycles, artificial oocyte activation (AOA) was applied and successfully rescued the fertilization failure and polyspermy phenotypes, with 31.3% (15/48) of the MII oocytes normally fertilized. However, 60.0% (9/15) of these normally fertilized zygotes were arrested at 2-5-cell stage, with one failing to cleave, indicating that PLCZ1 was not only necessary for fertilization, but also crucial for early embryonic development. However, these rescued zygotes showed a lower potential in developing into blastocysts when cultured in vitro. Thus, fresh cleavage transfer was tried and two live births were successfully achieved thereafter. In conclusion, this study provided novel mutations in PLCZ1 gene to expand the pathogenic mutational spectrum in male infertility and demonstrated that PLCZ1 was a crucial sperm-related genetic factor for early embryonic arrest. We also proposed that cleavage transfer after ICSI and AOA treatment could be a potential treatment method for male patients carrying bi-allelic mutations in PLCZ1.
Collapse
Affiliation(s)
- Yunying Lin
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yi Huang
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Boyu Li
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ting Zhang
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yichao Niu
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Shuanggang Hu
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ying Ding
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Guangxin Yao
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Zhe Wei
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ning Yao
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yejie Yao
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yao Lu
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yaqiong He
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Qinling Zhu
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ling Zhang
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
10
|
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: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [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
|
11
|
Kashir J, Mistry BV, BuSaleh L, Nomikos M, Almuqayyil S, Abu-Dawud R, AlYacoub N, Hamdan H, AlHassan S, Lai FA, Assiri AM, Coskun S. Antigen Unmasking Is Required to Clinically Assess Levels and Localisation Patterns of Phospholipase C Zeta in Human Sperm. Pharmaceuticals (Basel) 2023; 16:198. [PMID: 37259347 PMCID: PMC9962097 DOI: 10.3390/ph16020198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/10/2024] Open
Abstract
Mammalian oocyte activation is initiated by intracellular calcium (Ca2+) oscillations, driven by the testis-specific phospholipase C zeta (PLCζ). Sperm PLCζ analysis represents a diagnostic measure of sperm fertilisation capacity. The application of antigen unmasking/retrieval (AUM) generally enhanced the visualisation efficacy of PLCζ in mammalian sperm, but differentially affected the PLCζ profiles in sperm from different human males. It is unclear whether AUM affects the diagnosis of PLCζ in human sperm. Herein, we examined whether the application of AUM affected the correlation of PLCζ profiles with sperm parameters and fertilisation capacity. PLCζ fluorescence levels and localisation patterns were examined within the sperm of males undergoing fertility treatment (55 patients aged 29-53) using immunofluorescence in the absence/presence of AUM. The changes in PLCζ profiles following AUM were examined in relation to sperm health and fertilisation outcome. AUM enhanced the observable levels and specific localisation patterns of PLCζ in relation to both optimal sperm parameters and fertilisation outcome, without which significant differences were not observed. The extent of the change in levels and localisation ratios of PLCζ was also affected to a larger degree in terms of the optimal parameters of sperm fertility and fertilisation capacity by AUM. Collectively, AUM was essential to accurately assesses PLCζ in human sperm in both scientific and clinical contexts.
Collapse
Affiliation(s)
- Junaid Kashir
- Department of Biology, College of Arts and Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Bhavesh V. Mistry
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Lujain BuSaleh
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Michail Nomikos
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
| | - Sarah Almuqayyil
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Raed Abu-Dawud
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Nadya AlYacoub
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Saad AlHassan
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| | - F. Anthony Lai
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
| | - Abdullah M. Assiri
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Serdar Coskun
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11564, Saudi Arabia
| |
Collapse
|
12
|
Do follicle-stimulating hormone receptor polymorphisms in infertile men influence intracytoplasmic sperm injection outcomes? A prospective cohort study. ZYGOTE 2022; 30:809-818. [PMID: 36134751 DOI: 10.1017/s0967199422000302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The follicle-stimulating hormone (FSH) and its receptor regulate the quantity and quality of spermatozoa production. Several studies have analyzed the effect of single nucleotide polymorphisms (SNPs) in exon 10 of the FSH receptor (FSHR) on basic semen parameters without yet reaching a firm consensus. The aim of this study was to evaluate the effect of p.Thr307Ala and p.Asn680Ser polymorphisms in exon 10 of the FSHR gene, in infertile men, on intracytoplasmic sperm injection (ICSI) outcomes. This study was conducted between March 2019 and February 2020 on infertile couples undergoing ICSI at Al Hadi Laboratory and Medical Center, Lebanon. Couples with severe infertility factors that may impair gametogenesis/embryogenesis (e.g. advanced maternal age, premature ovarian failure, underwent gonadotoxic treatments, etc.) were excluded from the study. Semen and blood samples were collected from infertile men on the day of oocyte collection. Infertile men (n = 173) were screened for FSHR variants using polymerase chain reaction-restriction fragment length polymorphism. Moreover, fertilization rates, embryo quality, and pregnancy outcomes were evaluated. Higher sperm concentrations were found in the p.Thr307Ala group than the p.Thr307Thr (P < 0.01) and p.Ala307Ala (P < 0.05) groups. Furthermore, fertilization rate was significantly lower in the p.Ala307Ala genotype than in the p.Thr307Thr genotype (P < 0.05). We showed that FSHR variants in infertile men undergoing ICSI could affect sperm concentration, motility, and fertilization rates. Therefore, it will be important to confirm these results in further studies using a larger sample size.
Collapse
|
13
|
Marinaro J, Goldstein M. Microsurgical Management of Male Infertility: Compelling Evidence That Collaboration with Qualified Male Reproductive Urologists Enhances Assisted Reproductive Technology (ART) Outcomes. J Clin Med 2022; 11:jcm11154593. [PMID: 35956208 PMCID: PMC9369943 DOI: 10.3390/jcm11154593] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
A male factor plays a significant role in a couple's reproductive success. Today, advances in reproductive technology, such as intracytoplasmic sperm injection (ICSI), have allowed it to be possible for just a single sperm to fertilize an egg, thus, overcoming many of the traditional barriers to male fertility, such as a low sperm count, impaired motility, and abnormal morphology. Given these advances in reproductive technology, it has been questioned whether a reproductive urologist is needed for the evaluation and treatment of infertile and subfertile men. In this review, we aim to provide compelling evidence that collaboration between reproductive endocrinologists and reproductive urologists is essential for optimizing a couple's fertility outcomes, as well as for improving the health of infertile men and providing cost-effective care.
Collapse
Affiliation(s)
- Jessica Marinaro
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Marc Goldstein
- Center for Male Reproductive Medicine and Microsurgery, Weill Cornell Medicine, 525 East 68th St., Starr Pavilion, 9th Floor (Starr 900), New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
14
|
Mirsanei JS, Sheibak N, Zandieh Z, Mehdizadeh M, Aflatoonian R, Tabatabaei M, Mousavi AS, Amjadi F. Microfluidic chips as a method for sperm selection improve fertilization rate in couples with fertilization failure. Arch Gynecol Obstet 2022; 306:901-910. [PMID: 35650258 DOI: 10.1007/s00404-022-06618-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/07/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Sperm quality plays a vital role in successful fertilization and pregnancy. Patients with fertilization failure (total failure or low-fertilization rate) despite having normal semen parameters are a challenging group whose sperm cannot fertilize the oocyte via the intracytoplasmic sperm injection (ICSI) technique. Microfluidics is offered as a new method for proper sperm sorting. METHODS This study aimed to evaluate sperm parameters, DNA fragmentation index (DFI), expression of phospholipase C zeta 1 (PLCZ1), and transition nuclear proteins 1 (TNP1) mRNAs in sperm selected by microfluidic sperm sorting (MSS) chip compared with conventional density gradient centrifugation technique in patients with fertilization failure following ICSI. Subsequence fertilization rate and embryo quality were assayed. RESULTS Normal morphology and total motility were significantly higher, and DFI was significantly lower in sperm selected by the MSS chip in fertilization failure and control groups. The RT-PCR results demonstrated a significant increase in the expression of PLCZ1 and TNP1 genes in sperm of both groups selected by MSS chips compared to the DGC method. In addition, with the selected sperm by MSS chip, an increase in fertilization rate and improvement of embryo quality was obtained. CONCLUSION The present study findings show that sperm sorting by the microfluidic method improves fertilization rate in patients with poor fertilization outcomes following ICSI.
Collapse
Affiliation(s)
- Jamileh Sadat Mirsanei
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Nadia Sheibak
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandieh
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maryamsadat Tabatabaei
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Atieh Sadat Mousavi
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Amjadi
- Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran. .,Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Jones C, Meng X, Coward K. SPERM FACTORS AND EGG ACTIVATION: Phospholipase C zeta (PLCZ1) and the clinical diagnosis of oocyte activation deficiency. Reproduction 2022; 164:F53-F66. [PMID: 35312629 PMCID: PMC9175550 DOI: 10.1530/rep-21-0458] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/21/2022] [Indexed: 11/29/2022]
Abstract
Oocyte activation deficiency (OAD) remains the predominant cause of total/low fertilization rate in assisted reproductive technology. Phospholipase C zeta (PLCZ1) is the dominant sperm-specific factor responsible for triggering oocyte activation in mammals. OAD has been linked to numerous PLCZ1 abnormalities in patients experiencing failed in vitro fertilization or intracytoplasmic sperm injection cycles. While significant efforts have enhanced our understanding of the clinical relevance of PLCZ1, and the potential effects of genetic variants upon functionality, our ability to apply PLCZ1 in a diagnostic or therapeutic role remains limited. Artificial oocyte activation is the only option for patients experiencing OAD but lacks a reliable diagnostic approach. Immunofluorescence analysis has revealed that the levels and localization patterns of PLCZ1 within sperm can help us to indirectly diagnose a patient's ability to induce oocyte activation. Screening of the gene encoding PLCZ1 protein is also critical if we are to fully determine the extent to which genetic factors might play a role in the aberrant expression and/or localization patterns observed in infertile patients. Collectively, these findings highlight the clinical potential of PLCZ1, both as a prognostic indicator of OAD and eventually as a therapeutic agent. In this review, we focus on our understanding of the association between OAD and PLCZ1 by discussing the localization and expression of this key protein in human sperm, the potential genetic causes of OAD, and the diagnostic tools that are currently available to us to identify PLCZ1 deficiency and select patients that would benefit from targeted therapy.
Collapse
Affiliation(s)
- C Jones
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Headington, Oxford, UK
| | - X Meng
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Headington, Oxford, UK
| | - K Coward
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Women’s Centre, John Radcliffe Hospital, Headington, Oxford, UK
| |
Collapse
|
16
|
Shafqat A, Kashir J, Alsalameh S, Alkattan K, Yaqinuddin A. Fertilization, Oocyte Activation, Calcium Release and Epigenetic Remodelling: Lessons From Cancer Models. Front Cell Dev Biol 2022; 10:781953. [PMID: 35309905 PMCID: PMC8931327 DOI: 10.3389/fcell.2022.781953] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022] Open
Abstract
Oocyte activation deficiency (OAD) is the basis of Total Fertilisation Failure (TFF) and is attributed to mutations in the PLCζ gene—termed male factor infertility. This derives abnormal Ca2+ oscillations and could be the main cause of primary disruptions in the gene expression of Ca2+-related proteins. Epigenetic mechanisms are universally accepted as key regulators of gene expression. However, epigenetic dysregulations have not been considered as potential mechanisms of oocyte-borne OAD. Herein, we discuss changes in the DNA methylome during oogenesis and embryogenesis. We further highlight key pathways comprising the oocyte Ca2+ toolkit, which could be targets of epigenetic alterations, especially aberrations in DNA methylation. Considering that the vast majority of epigenetic modifications examined during fertilization revolve around alterations in DNA methylation, we aim in this article to associate Ca2+-specific mechanisms with these alterations. To strengthen this perspective, we bring evidence from cancer research on the intricate link between DNA methylation and Ca2+ signaling as cancer research has examined such questions in a lot more detail. From a therapeutic standpoint, if our hypothesis is proven to be correct, this will explain the cause of TFF in idiopathic cases and will open doors for novel therapeutic targets.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- *Correspondence: Ahmed Yaqinuddin,
| |
Collapse
|
17
|
Recurrent spontaneous oocyte activation causes female infertility. J Assist Reprod Genet 2022; 39:675-680. [PMID: 35156150 PMCID: PMC8995232 DOI: 10.1007/s10815-022-02435-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/09/2022] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Spontaneous oocyte activation (SOA) is a recently classified phenomenon characterized by the presence of a single pronucleus immediately following oocyte retrieval, without the apparent involvement of sperm. SOA currently remains poorly understood in humans, with no clear genetic or pathological factor(s). Herein, we report two separate cases of recurrent spontaneous oocyte activation, investigating potential avenues to identify causative etiology. METHODS Two patients with several cycles with SOA have undergone further genetic and embryologic investigation to reveal underlying causes for SOA and provide a treatment if possible. RESULTS One case was a patient with recurrent pregnancy loss and the other was diagnosed as unexplained infertility. In the first case, 61 out of 69 oocytes retrieved exhibited SOA in five cycles while in the second case 44 out of 49 oocytes exhibited SOA in five cycles. Oocytes were injected with sperm; embryo development and presence of paternal contribution were investigated. No pregnancy is ensued following embryo transfer in both patients. Time-lapse imaging of embryogenesis from the second case did not reveal even momentary second pronucleus appearance. We also performed clinical whole exome sequencing for both patients but did not identify any disease-causing variant. CONCLUSION Patients with SOA suffer from infertility. Our results indicate that more investigation is required to understand the etiology of SOA in humans concentrating on the molecular mechanisms that underpin regulation of oocyte activation and calcium dynamics need to be investigated to fully understand, and perhaps in the future rectify, recurrent SOA.
Collapse
|
18
|
Kashir J, Ganesh D, Jones C, Coward K. OUP accepted manuscript. Hum Reprod Open 2022; 2022:hoac003. [PMID: 35261925 PMCID: PMC8894871 DOI: 10.1093/hropen/hoac003] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/16/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Oocyte activation deficiency (OAD) is attributed to the majority of cases underlying failure of ICSI cycles, the standard treatment for male factor infertility. Oocyte activation encompasses a series of concerted events, triggered by sperm-specific phospholipase C zeta (PLCζ), which elicits increases in free cytoplasmic calcium (Ca2+) in spatially and temporally specific oscillations. Defects in this specific pattern of Ca2+ release are directly attributable to most cases of OAD. Ca2+ release can be clinically mediated via assisted oocyte activation (AOA), a combination of mechanical, electrical and/or chemical stimuli which artificially promote an increase in the levels of intra-cytoplasmic Ca2+. However, concerns regarding safety and efficacy underlie potential risks that must be addressed before such methods can be safely widely used. OBJECTIVE AND RATIONALE Recent advances in current AOA techniques warrant a review of the safety and efficacy of these practices, to determine the extent to which AOA may be implemented in the clinic. Importantly, the primary challenges to obtaining data on the safety and efficacy of AOA must be determined. Such questions require urgent attention before widespread clinical utilization of such protocols can be advocated. SEARCH METHODS A literature review was performed using databases including PubMed, Web of Science, Medline, etc. using AOA, OAD, calcium ionophores, ICSI, PLCζ, oocyte activation, failed fertilization and fertilization failure as keywords. Relevant articles published until June 2019 were analysed and included in the review, with an emphasis on studies assessing large-scale efficacy and safety. OUTCOMES Contradictory studies on the safety and efficacy of AOA do not yet allow for the establishment of AOA as standard practice in the clinic. Heterogeneity in study methodology, inconsistent sample inclusion criteria, non-standardized outcome assessments, restricted sample size and animal model limitations render AOA strictly experimental. The main scientific concern impeding AOA utilization in the clinic is the non-physiological method of Ca2+ release mediated by most AOA agents, coupled with a lack of holistic understanding regarding the physiological mechanism(s) underlying Ca2+ release at oocyte activation. LIMITATIONS, REASONS FOR CAUTION The number of studies with clinical relevance using AOA remains significantly low. A much wider range of studies examining outcomes using multiple AOA agents are required. WIDER IMPLICATIONS In addition to addressing the five main challenges of studies assessing AOA safety and efficacy, more standardized, large-scale, multi-centre studies of AOA, as well as long-term follow-up studies of children born from AOA, would provide evidence for establishing AOA as a treatment for infertility. The delivery of an activating agent that can more accurately recapitulate physiological fertilization, such as recombinant PLCζ, is a promising prospect for the future of AOA. Further to PLCζ, many other avenues of physiological oocyte activation also require urgent investigation to assess other potential physiological avenues of AOA. STUDY FUNDING/COMPETING INTERESTS D.G. was supported by Stanford University’s Bing Overseas Study Program. J.K. was supported by a Healthcare Research Fellowship Award (HF-14-16) made by Health and Care Research Wales (HCRW), alongside a National Science, Technology, and Innovation plan (NSTIP) project grant (15-MED4186-20) awarded by the King Abdulaziz City for Science and Technology (KACST). The authors have no competing interests to declare.
Collapse
Affiliation(s)
| | | | - Celine Jones
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Oxford, UK
| | - Kevin Coward
- Correspondence address. Nuffield Department of Women’s & Reproductive Health, University of Oxford, Level 3, Women’s Centre, John Radcliffe Hospital, Oxford, OS3 9DU, UK. E-mail: https://orcid.org/0000-0003-3577-4041
| |
Collapse
|
19
|
Xue Y, Cheng X, Xiong Y, Li K. Gene mutations associated with fertilization failure after in vitro fertilization/intracytoplasmic sperm injection. Front Endocrinol (Lausanne) 2022; 13:1086883. [PMID: 36589837 PMCID: PMC9800785 DOI: 10.3389/fendo.2022.1086883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Fertilization failure during assisted reproductive technologies (ART) is often unpredictable, as this failure is encountered only after in vitro fertilization (IVF) and intracytoplasmic sperm injection (ICSI) have been performed. The etiology of fertilization failure remains elusive. More and more mutations of genes are found to be involved in human fertilization failure in infertile patients as high throughput sequencing techniques are becoming widely applied. In this review, the mutations of nine important genes expressed in sperm or oocytes, PLCZ1, ACTL7A, ACTL9, DNAH17, WEE2, TUBB8, NLRP5, ZP2, and TLE6, were summarized and discussed. These abnormalities mainly have shown Mendelian patterns of inheritance, including dominant and recessive inheritance, although de novo mutations were present in some cases. The review revealed the crucial roles of each reported gene in the fertilization process and summarized all known mutations and their corresponding phenotypes. The review suggested the mutations might become promising targets for precision treatments in reproductive medicine. Moreover, our work will provide some helpful clues for genetic counseling, risk prediction, and optimizing clinical treatments for human infertility by supplying the useful and timely information on the genetic causes leading to fertilization failure.
Collapse
Affiliation(s)
- Yamei Xue
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohong Cheng
- Institute for Reproductive Health, School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Yuping Xiong
- Institute for Reproductive Health, School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Kun Li
- Institute for Reproductive Health, School of Pharmacy, Hangzhou Medical College, Hangzhou, China
- Zhejiang Provincial Laboratory of Experimental Animal’s & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Kun Li,
| |
Collapse
|
20
|
Aras-Tosun D, Cakar Z, Can A, Ozkavukcu S, Kaplanoglu I, Cinar O. Phospholipase C-zeta levels are not correlated with fertilisation rates in infertile couples. Andrologia 2021; 54:e14269. [PMID: 34651330 DOI: 10.1111/and.14269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/29/2022] Open
Abstract
In mammals, 'oocyte activation' is triggered by certain proteins, one of which is phospholipase C-zeta. Recent evidence suggests that low expression of phospholipase C-zeta might be associated with male infertility, while a limited number of studies claimed the opposite. This study was designed to test whether quantity of phospholipase C-zeta and in vitro fertilisation rates are correlated or not, assessed by flow cytometry. Semen samples from 43 infertile couples were analysed for the percentage and mean fluorescent intensity (MFI) of phospholipase C-zeta protein. Results were confirmed by immunofluorescent labelling. Patients with a fertilisation rate of 40% or lower were involved in the low fertilisation group, while the high fertilization group consisted of patients with a fertilisation rate of 60% and higher. Quantitative analyses by flow cytometry showed no significant difference among the low fertilisation and high fertilisation groups when phospholipase C-zeta ratio or MFI was considered. No correlation was found between pregnancy rates and phospholipase C-zeta quantity. None of the total fertilisation failure cases were lack of phospholipase C-zeta. In fact, fertilisation was possible even when phospholipase C-zeta levels were very low. Thus, we concluded that phospholipase C-zeta quantity cannot be considered as a diagnostic tool for male infertility.
Collapse
Affiliation(s)
- Duru Aras-Tosun
- Department of Histology and Embryology, Ankara University School of Dentistry, Ankara, Turkey
| | - Zeynep Cakar
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Alp Can
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Sinan Ozkavukcu
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey.,Centre for Assisted Reproduction, Ankara University School of Medicine, Ankara, Turkey
| | - Iskender Kaplanoglu
- Centre for Assisted Reproductive Medicine, TCSB Etlik Zubeyde Hanim Women's Health Teaching and Research Hospital, Ankara, Turkey
| | - Ozgur Cinar
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey.,Centre for Assisted Reproduction, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
21
|
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: 4] [Impact Index Per Article: 1.0] [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
|
22
|
Diagnosis and Treatment of Male Infertility-Related Fertilization Failure. J Clin Med 2020; 9:jcm9123899. [PMID: 33271815 PMCID: PMC7761017 DOI: 10.3390/jcm9123899] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022] Open
Abstract
Infertility affects approximately 15% of reproductive-aged couples worldwide, of which up to 30% of the cases are caused by male factors alone. The origin of male infertility is mostly attributed to sperm abnormalities, of which many are caused by genetic defects. The development of intracytoplasmic sperm injection (ICSI) has helped to circumvent most male infertility conditions. However, there is still a challenging group of infertile males whose sperm, although having normal sperm parameters, are unable to activate the oocyte, even after ICSI treatment. While ICSI generally allows fertilization rates of 70 to 80%, total fertilization failure (FF) still occurs in 1 to 3% of ICSI cycles. Phospholipase C zeta (PLCζ) has been demonstrated to be a critical sperm oocyte activating factor (SOAF) and the absence, reduced, or altered forms of PLCζ have been shown to cause male infertility-related FF. The purpose of this review is to (i) summarize the current knowledge on PLCζ as the critical sperm factor for successful fertilization, as well as to discuss the existence of alternative sperm-induced oocyte activation mechanisms, (ii) describe the diagnostic tests available to determine the cause of FF, and (iii) summarize the beneficial effect of assisted oocyte activation (AOA) to overcome FF.
Collapse
|