1
|
Sheahan TD, Warwick CA, Cui AY, Baranger DAA, Perry VJ, Smith KM, Manalo AP, Nguyen EK, Koerber HR, Ross SE. Kappa opioids inhibit spinal output neurons to suppress itch. SCIENCE ADVANCES 2024; 10:eadp6038. [PMID: 39321286 PMCID: PMC11423883 DOI: 10.1126/sciadv.adp6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
Itch is a protective sensation that drives scratching. Although specific cell types have been proposed to underlie itch, the neural basis for itch remains unclear. Here, we used two-photon Ca2+ imaging of the dorsal horn to visualize neuronal populations that are activated by itch-inducing agents. We identify a convergent population of spinal interneurons recruited by diverse itch-causing stimuli that represents a subset of neurons that express the gastrin-releasing peptide receptor (GRPR). Moreover, we find that itch is conveyed to the brain via GRPR-expressing spinal output neurons that target the lateral parabrachial nuclei. We then show that the kappa opioid receptor agonist nalfurafine relieves itch by selectively inhibiting GRPR spinoparabrachial neurons. These experiments provide a population-level view of the spinal neurons that respond to pruritic stimuli, pinpoint the output neurons that convey itch to the brain, and identify the cellular target of kappa opioid receptor agonists for the inhibition of itch.
Collapse
Affiliation(s)
- Tayler D Sheahan
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles A Warwick
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abby Y Cui
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A A Baranger
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Vijay J Perry
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kelly M Smith
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison P Manalo
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eileen K Nguyen
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - H Richard Koerber
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah E Ross
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Gonzalez-Castro RA, Whitcomb LA, Pinsinski EC, Carnevale EM. Cryopreservation of equine spermatozoa reduces plasma membrane integrity and phospholipase C zeta 1 content as associated with oocyte activation. Andrology 2024; 12:918-931. [PMID: 37608516 DOI: 10.1111/andr.13517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 07/21/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Phospholipase C zeta (PLCZ1) is considered the major sperm-borne oocyte activation factor. Cryopreserved stallion spermatozoa are commonly used for intracytoplasmic sperm injection (ICSI). However, plasma membrane damage and protein modifications caused by cryopreservation could impair sperm structure and function, leading to a reduction of PLCZ1 and oocyte activation after ICSI. OBJECTIVES We compared membrane integrity and PLCZ1 abundance in populations for fresh, frozen, and refrozen stallion spermatozoa, either thawed and refrozen at room or low temperature; and examined the effect of relative PLCZ1 content on cleavage after ICSI. MATERIALS AND METHODS Western blotting, ELISA, and immunofluorescence were conducted in stallion spermatozoa, freezing extenders, and detergent-extracted sperm fractions to detect and quantify PLCZ1. Retrospectively, PLCZ1 content and cleavage rate were analyzed. Fresh, frozen, and refrozen at room and low temperatures spermatozoa were evaluated for acrosomal and plasma membrane integrity and PLCZ1 content using flow cytometry. RESULTS Western blotting, ELISA, and immunofluorescence revealed significant reduction of PLCZ1 in spermatozoa after cryopreservation and confirmed PLCZ1 detection in extenders. After detergent extraction, a PLCZ1-nonextractable fraction remained in the postacrosomal region of spermatozoa. Plasma membrane integrity was significantly reduced after freezing. Acrosomal and plasma membrane integrity were similar between frozen and refrozen samples at low temperature, but both were significantly higher than samples refrozen at room temperature. Acrosomal and plasma membrane integrity significantly correlated to PLCZ1 content. Percentages of PLCZ1-labeled spermatozoa and PLCZ1 content were reduced after freezing but not after refreezing. Relative content and localization of PLCZ1 were associated with cleavage rates after ICSI. DISCUSSION AND CONCLUSION Sperm PLCZ1 content associates with cleavage rates after ICSI. Cryopreservation is detrimental to sperm plasma membrane integrity and PLCZ1 retention. However, refreezing did not result in additional PLCZ1 loss. Refreezing stallion spermatozoa at a low temperature resulted in better survival but did not improve PLCZ1 retention.
Collapse
Affiliation(s)
- Raul A Gonzalez-Castro
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Luke A Whitcomb
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Emma C Pinsinski
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Elaine M Carnevale
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
3
|
Montjean D, Beaumont M, Natiq A, Louanjli N, Hazout A, Miron P, Liehr T, Cabry R, Ratbi I, Benkhalifa M. Genome and Epigenome Disorders and Male Infertility: Feedback from 15 Years of Clinical and Research Experience. Genes (Basel) 2024; 15:377. [PMID: 38540436 PMCID: PMC10970370 DOI: 10.3390/genes15030377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 06/14/2024] Open
Abstract
Infertility affects around 20% of couples of reproductive age; however, in some societies, as many as one-third of couples are unable to conceive. Different factors contribute to the decline of male fertility, such us environmental and professional exposure to endocrine disruptors, oxidative stress, and life habits with the risk of de novo epigenetics dysregulation. Since the fantastic development of new "omes and omics" technologies, the contribution of inherited or de novo genomes and epigenome disorders to male infertility have been further elucidated. Many other techniques have become available to andrology laboratories for the investigation of genome and epigenome integrity and the maturation and the competency of spermatozoa. All these new methods of assessment are highlighting the importance of genetics and epigenetics investigation for assisted reproduction pathology and for supporting professionals in counselling patients and proposing different management strategies for male infertility. This aims to improve clinical outcomes while minimizing the risk of genetics or health problems at birth.
Collapse
Affiliation(s)
- Debbie Montjean
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
| | - Marion Beaumont
- Genetics Department, Eylau/Unilabs Laboratory, 92110 Clichy, France;
| | - Abdelhafid Natiq
- Center for Genomics of Human Pathologies (GENOPATH), Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco (I.R.)
- National Laboratory Mohammed VI, Mohammed VI Foundation of Casablanca, Casablanca, Morocco
| | | | - Andre Hazout
- Andro-Genetics Unit, Labomac, Casablanca, Morocco (A.H.)
| | - Pierre Miron
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
| | - Thomas Liehr
- Institute für Humangenetik, Universitätsklinikum Jena, Friedrich Schiller Universität, 07743 Jena, Germany
| | - Rosalie Cabry
- Reproductive Medicine, Reproductive Biology & Genetics, CECOS Picardie, University Hospital & School of Medicine, Picardie University Jules Verne, 80000 Amiens, France
- PeriTox Laboratory, Perinatality & Toxic Risks, UMR-I 01 INERIS, Picardie University Jules Verne, 80000 Amiens, France
| | - Ilham Ratbi
- Center for Genomics of Human Pathologies (GENOPATH), Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco (I.R.)
- Medical Genetics Unit, Ibn Sina University Hospital Center, Rabat, Morocco
| | - Moncef Benkhalifa
- Fertilys Fertility Centers Laval and Brossard, 1950 Maurice-Gauvin Street, Laval, QC H7S 1Z5, Canada; (D.M.)
- Reproductive Medicine, Reproductive Biology & Genetics, CECOS Picardie, University Hospital & School of Medicine, Picardie University Jules Verne, 80000 Amiens, France
- PeriTox Laboratory, Perinatality & Toxic Risks, UMR-I 01 INERIS, Picardie University Jules Verne, 80000 Amiens, France
| |
Collapse
|
4
|
Gonzalez-Castro RA, Carnevale EM. Phospholipase C Zeta 1 (PLCZ1): The Function and Potential for Fertility Assessment and In Vitro Embryo Production in Cattle and Horses. Vet Sci 2023; 10:698. [PMID: 38133249 PMCID: PMC10747197 DOI: 10.3390/vetsci10120698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Phospholipase C Zeta 1 (PLCZ1) is considered a major sperm-borne oocyte activation factor. After gamete fusion, PLCZ1 triggers calcium oscillations in the oocyte, resulting in oocyte activation. In assisted fertilization, oocyte activation failure is a major cause of low fertility. Most cases of oocyte activation failures in humans related to male infertility are associated with gene mutations and/or altered PLCZ1. Consequently, PLCZ1 evaluation could be an effective diagnostic marker and predictor of sperm fertilizing potential for in vivo and in vitro embryo production. The characterization of PLCZ1 has been principally investigated in men and mice, with less known about the PLCZ1 impact on assisted reproduction in other species, such as cattle and horses. In horses, sperm PLCZ1 varies among stallions, and sperm populations with high PLCZ1 are associated with cleavage after intracytoplasmic sperm injection (ICSI). In contrast, bull sperm is less able to initiate calcium oscillations and undergo nuclear remodeling, resulting in poor cleavage after ICSI. Advantageously, injections of PLCZ1 are able to rescue oocyte failure in mouse oocytes after ICSI, promoting full development and birth. However, further research is needed to optimize PLCZ1 diagnostic tests for consistent association with fertility and to determine whether PLCZ1 as an oocyte-activating treatment is a physiological, efficient, and safe method for improving assisted fertilization in cattle and horses.
Collapse
Affiliation(s)
| | - Elaine M. Carnevale
- Equine Reproduction Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA;
| |
Collapse
|
5
|
Sheahan TD, Warwick CA, Cui AY, Baranger DA, Perry VJ, Smith KM, Manalo AP, Nguyen EK, Koerber HR, Ross SE. Identification of a convergent spinal neuron population that encodes itch. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560205. [PMID: 37873278 PMCID: PMC10592866 DOI: 10.1101/2023.09.29.560205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Itch is a protective sensation that drives scratching. Although specific cell types have been proposed to underlie itch, the neural circuit basis for itch remains unclear. Here, we used two-photon Ca2+ imaging of the dorsal horn to visualize the neuronal populations that are activated by itch-inducing agents. We identify a convergent population of spinal neurons that is defined by the expression of GRPR. Moreover, we discover that itch is conveyed to the brain via GRPR-expressing spinal output neurons that target the lateral parabrachial nucleus. Further, we show that nalfurafine, a clinically effective kappa opioid receptor agonist, relieves itch by inhibiting GRPR spinoparabrachial neurons. Finally, we demonstrate that a subset of GRPR spinal neurons show persistent, cell-intrinsic Ca2+ oscillations. These experiments provide the first population-level view of the spinal neurons that respond to pruritic stimuli, pinpoint the output neurons that convey itch to the brain, and identify the cellular target of kappa opioid receptor agonists for the inhibition of itch.
Collapse
Affiliation(s)
- Tayler D. Sheahan
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Co-first authors
| | - Charles A. Warwick
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Co-first authors
| | - Abby Y. Cui
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David A.A. Baranger
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis Missouri, USA
| | - Vijay J. Perry
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kelly M. Smith
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Current Address: Biohaven Pharmaceuticals, LTD, Pittsburgh, Pennsylvania, USA
| | - Allison P. Manalo
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Eileen K. Nguyen
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Current Address: Department of Anesthesiology and Perioperative Care, University of California, Los Angeles, Los Angeles, California, USA
| | - H. Richard Koerber
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sarah E. Ross
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Lead contact
| |
Collapse
|
6
|
Nguyen HT, Do SQ, Athurupana R, Wakai T, Funahashi H. Rapid thawing of frozen bull spermatozoa by transient exposure to 70 °C improves the viability, motility and mitochondrial health. Anim Reprod 2023; 20:e20220127. [PMID: 38026001 PMCID: PMC10681132 DOI: 10.1590/1984-3143-ar2022-0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 07/13/2023] [Indexed: 12/01/2023] Open
Abstract
Up to now, the definitive conclusion of the positive effects of rapid transient thawing at higher temperatures for shorter durations has not been obtained yet and is still under discussion due to some contradictory findings and limited assessment of post-thawed parameters. The purpose of the current study was to evaluate the effectiveness of rapid thawing in water at 70 °C by using various post-thawed parameters of frozen bull spermatozoa. Experiment 1, monitoring the change of temperature inside frozen bull straw thawed in water at different temperatures. Experiment 2, evaluation of various post-thawed characteristics of frozen bull spermatozoa thawed in water at different temperatures by using a computer-assisted sperm analysis, flow cytometry and immunocytochemistry. The time it took for the temperature inside the straw to warm up to 15 °C was nearly twice as faster when the straw was thawed in 70 °C water compared with 39 °C. Although there were differences among bulls, viability, motility, and mitochondrial membrane potential of spermatozoa thawed at 70 °C for 8 seconds and stabilized at 39 °C for 52 seconds were significantly higher than those of controls (thawed at 39 °C for 60 seconds) at 0 and 3 h after thawing. Just after thawing, however, there were no differences in acrosome integrity and distribution of phospholipase C zeta1, whereas mitochondrial reactive oxygen species production was significantly lower in spermatozoa thawed at 70 °C. From these results, we conclude that rapid thawing at 70 °C and then stabilization at 39 °C significantly improves viability, motility and mitochondrial health of bull spermatozoa rather than conventional thawing at 39 °C. The beneficial effect of rapid transient thawing could be due to shorter exposure to temperatures outside the physiological range, consequently maintaining mitochondrial health.
Collapse
Affiliation(s)
- Hai Thanh Nguyen
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
- Department of Animal Production, Faculty of Animal Science and Veterinary Medicine, Nong Lam University, Ho Chi Minh City, Vietnam
| | - Son Quang Do
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Rukmali Athurupana
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Takuya Wakai
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Hiroaki Funahashi
- Department of Animal Science, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| |
Collapse
|
7
|
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: 1.0] [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
|
8
|
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: 2.0] [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
|
9
|
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: 2.0] [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
|
10
|
Huang G, Zhang X, Yao G, Huang L, Wu S, Li X, Guo J, Wen Y, Wang Y, Shang L, Li N, Xu W. A loss-of-function variant in SSFA2 causes male infertility with globozoospermia and failed oocyte activation. Reprod Biol Endocrinol 2022; 20:103. [PMID: 35836265 PMCID: PMC9281110 DOI: 10.1186/s12958-022-00976-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
Globozoospermia (OMIM: 102530) is a rare type of teratozoospermia (< 0.1%). The etiology of globozoospermia is complicated and has not been fully revealed. Here, we report an infertile patient with globozoospermia. Variational analysis revealed a homozygous missense variant in the SSFA2 gene (NM_001130445.3: c.3671G > A; p.R1224Q) in the patient. This variant significantly reduced the protein expression of SSFA2. Immunofluorescence staining showed positive SSFA2 expression in the acrosome of human sperm. Liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) and Coimmunoprecipitation (Co-IP) analyses identified that GSTM3 and Actin interact with SSFA2. Further investigation revealed that for the patient, regular intracytoplasmic sperm injection (ICSI) treatment had a poor prognosis. However, Artificial oocyte activation (AOA) by a calcium ionophore (A23187) after ICSI successfully rescued the oocyte activation failure for the patient with the SSFA2 variant, and the couple achieved a live birth. This study revealed that SSFA2 plays an important role in acrosome formation, and the homozygous c.3671G > A loss-of-function variant in SSFA2 caused globozoospermia. SSFA2 may represent a new gene in the genetic diagnosis of globozoospermia, especially the successful outcome of AOA-ICSI treatment for couples, which has potential value for clinicians in their treatment regimen selections.
Collapse
Affiliation(s)
- Gelin Huang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Joint Lab for Reproductive Medicine(SCU-CUHK), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xueguang Zhang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Joint Lab for Reproductive Medicine(SCU-CUHK), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Guanping Yao
- Department of Reproductive Medicine Center, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lin Huang
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Joint Lab for Reproductive Medicine(SCU-CUHK), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Sixian Wu
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Joint Lab for Reproductive Medicine(SCU-CUHK), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaoliang Li
- Department of Reproductive Endocrinology of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Juncen Guo
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Joint Lab for Reproductive Medicine(SCU-CUHK), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuting Wen
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Joint Lab for Reproductive Medicine(SCU-CUHK), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Wang
- Department of Reproductive Endocrinology of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Lijun Shang
- School of Human Sciences, London Metropolitan University, London, UK
| | - Na Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Wenming Xu
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Joint Lab for Reproductive Medicine(SCU-CUHK), West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Shan Y, Zhao H, Zhao D, Wang J, Cui Y, Bao H. Assisted Oocyte Activation With Calcium Ionophore Improves Pregnancy Outcomes and Offspring Safety in Infertile Patients: A Systematic Review and Meta-Analysis. Front Physiol 2022; 12:751905. [PMID: 35140624 PMCID: PMC8819094 DOI: 10.3389/fphys.2021.751905] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/21/2021] [Indexed: 12/21/2022] Open
Abstract
This study aimed to evaluate the efficacy and safety of calcium ionophore during assisted oocyte activation (AOA). This meta-analysis contained randomized controlled trials and prospective observational and retrospective trials. The summary odds ratio (OR) with 95% confidence intervals (CIs) was calculated for clinical pregnancy rate and live birth rate. Both fixed and random effects models were applied. A total of 22 studies were included into this meta-analysis. Seventeen of the included studies showed that calcium ionophore increased the clinical pregnancy rate (OR, 2.14; 95% CI, 1.38–3.31). Similarly, 14 studies indicated that AOA with calcium ionophore during intracytoplasmic sperm injection (ICSI) improved the live birth rate considerably (OR, 2.65; 95% CI, 1.53–4.60). Moreover, fertilization, blastocyst formation, and implantation rate were higher after using AOA with calcium ionophore combined with ICSI. In addition, calcium ionophore did not increase top-quality embryo rate, cleavage rate, miscarriage rate, congenital birth defects, and neonatal sex ratio. Therefore, calcium ionophore followed by ICSI not only significantly improved live birth and overall pregnancy, but also did not affect the incidence of miscarriage, congenital birth defects, and neonatal sex ratio. This meta-analysis indicated that using calcium ionophore to activate oocytes was beneficial for couples with poor fertilization rates following ICSI.
Collapse
|
12
|
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: 22] [Impact Index Per Article: 11.0] [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
|
13
|
Wang A, Huang S, Liu M, Wang B, Wu F, Zhu D, Zhao X. Clinical exome sequencing identifies novel compound heterozygous mutations of the WEE2 gene in primary infertile women with fertilization failure. Gynecol Endocrinol 2021; 37:1096-1101. [PMID: 33904356 DOI: 10.1080/09513590.2021.1916458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE The genetic basis of fertilization failure after intracytoplasmic sperm injection (ICSI) is largely unknown and the aim of this study is to investigate the genetic causes of fertilization failure in primary infertile women. METHODS Six affected women diagnosed with infertility and fertilization failure were recruited. The genetically pathogenic factor of their fertilization failures were investigated by clinical exome sequencing. One hundred healthy controls were verified by Sanger sequencing. RESULTS Novel compound heterozygous mutations c.625G > T and c.759-2A > G of WEE2 in one affected individual were revealed by clinical exome sequencing. Trios analysis of the mutations represented an autosomal recessive pattern. The nonsense mutation c.625G > T (p.Glu209*) indicated the truncation of the WEE2 protein and c.759-2A > G was predicted to affect the splicing. CONCLUSIONS The novel variants extend the spectrum of WEE2 mutations, which promotes the prognostic value of testing for WEE2 mutations in infertile women with fertilization failure.
Collapse
Affiliation(s)
- Ancong Wang
- Department of Reproductive Medicine, Linyi People's Hospital, Linyi, PR China
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, PR China
| | - Shan Huang
- Department of Respiratory Medicine, Linyi Hospital of traditional Chinese Medicine, Linyi, PR China
| | - Min Liu
- Department of Reproductive Medicine, Linyi People's Hospital, Linyi, PR China
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, PR China
| | - Baosong Wang
- Department of Reproductive Medicine, Linyi People's Hospital, Linyi, PR China
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, PR China
| | - Fengxia Wu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Shandong University, Jinan, PR China
| | - Dongyi Zhu
- Department of Reproductive Medicine, Linyi People's Hospital, Linyi, PR China
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, PR China
| | - Xiangyu Zhao
- Department of Medical Genetics, Linyi People's Hospital, Linyi, PR China
| |
Collapse
|
14
|
Ferrer-Buitrago M, Tilleman L, Thys V, Hachem A, Boel A, Van Nieuwerburgh F, Deforce D, Leybaert L, De Sutter P, Parrington J, Heindryckx B. Comparative study of preimplantation development following distinct assisted oocyte activation protocols in a PLC-zeta knockout mouse model. Mol Hum Reprod 2021; 26:801-815. [PMID: 32898251 DOI: 10.1093/molehr/gaaa060] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
Mammalian fertilization encompasses a series of Ca2+ oscillations initiated by the sperm factor phospholipase C zeta (PLCζ). Some studies have shown that altering the Ca2+ oscillatory regime at fertilization affects preimplantation blastocyst development. However, assisted oocyte activation (AOA) protocols can induce oocyte activation in a manner that diverges profoundly from the physiological Ca2+ profiling. In our study, we used the newly developed PLCζ-null sperm to investigate the independent effect of AOA on mouse preimplantation embryogenesis. Based on previous findings, we hypothesized that AOA protocols with Ca2+ oscillatory responses might improve blastocyst formation rates and differing Ca2+ profiles might alter blastocyst transcriptomes. A total of 326 MII B6D2F1-oocytes were used to describe Ca2+ profiles and to compare embryonic development and individual blastocyst transcriptomes between four control conditions: C1 (in-vivo fertilization), C2 (ICSI control sperm), C3 (parthenogenesis) and C4 (ICSI-PLCζ-KO sperm) and four AOA groups: AOA1 (human recombinant PLCζ), AOA2 (Sr2+), AOA3 (ionomycin) and AOA4 (TPEN). All groups revealed remarkable variations in their Ca2+ profiles; however, oocyte activation rates were comparable between the controls (91.1% ± 13.8%) and AOA (86.9% ± 11.1%) groups. AOA methods which enable Ca2+ oscillatory responses (AOA1: 41% and AOA2: 75%) or single Ca2+ transients (AOA3: 50%) showed no significantly different blastocyst rates compared to ICSI control group (C2: 70%). In contrast, we observed a significant decrease in compaction (53% vs. 83%) and blastocyst rates (41% vs. 70%) in the absence of an initial Ca2+ trigger (AOA4) compared with the C2 group. Transcription profiles did not identify significant differences in gene expression levels between the ICSI control group (C2) and the four AOA groups.
Collapse
Affiliation(s)
- M Ferrer-Buitrago
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium.,CREA. Medicina de la Reproducción S.L. Calle San Martín, 4 - 46003 (Valencia, Spain)
| | - L Tilleman
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - V Thys
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - A Hachem
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.,Department of Anatomy, College of Veterinary Medicine, University of Al-Qadisiyah, Diwaniyah City, Iraq
| | - A Boel
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - F Van Nieuwerburgh
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - D Deforce
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - L Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - P De Sutter
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - J Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - B Heindryckx
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
15
|
Mu J, Zhang Z, Wu L, Fu J, Chen B, Yan Z, Li B, Zhou Z, Wang W, Zhao L, Dong J, Kuang Y, Sun X, He L, Wang L, Sang Q. The identification of novel mutations in PLCZ1 responsible for human fertilization failure and a therapeutic intervention by artificial oocyte activation. Mol Hum Reprod 2021; 26:80-87. [PMID: 31953539 DOI: 10.1093/molehr/gaaa003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Fertilization involves a series of molecular events immediately following egg-sperm fusion; Ca2+ oscillations are the earliest signaling event, and they initiate the downstream reactions including pronucleus formation. Successful human reproduction requires normal fertilization. In clinical IVF or ICSI attempts, some infertile couples suffer from recurrent fertilization failure. However, the genetic reasons for fertilization failure are largely unknown. Here, we recruited several couples diagnosed with fertilization failure even though their gametes are morphologically normal. Through whole-exome sequencing and Sanger sequencing, we identified biallelic mutations in gene-encoding phospholipase C zeta 1 (PLCZ1) in four independent males in couples diagnosed with fertilization failure. Western blotting showed that missense mutations decreased the level of PLCZ1 and that nonsense or frameshift mutations resulted in undetectable or truncated proteins. Expression of these mutations in mice significantly reduced the levels of oocyte activation. Artificial oocyte activation in patient oocytes could rescue the phenotype of fertilization failure and help establish pregnancy and lead to live birth. Our findings expand the spectrum of PLCZ1 mutations that are responsible for human fertilization failure and provide a potentially feasible therapeutic treatment for these patients.
Collapse
Affiliation(s)
- Jian Mu
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032
| | - Zhihua Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032
| | - Ling Wu
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Biaobang Chen
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032
| | - Zheng Yan
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Bin Li
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Zhou Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032
| | - Wenjing Wang
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032
| | - Lin Zhao
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032
| | - Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032
| | - Yanping Kuang
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032.,Zhuhai Fudan Innovation Institute, Zhuhai, Guangdong 519000, China.,Shanghai Center for Women and Children's Health, Shanghai, 200062, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032.,Zhuhai Fudan Innovation Institute, Zhuhai, Guangdong 519000, China
| |
Collapse
|
16
|
Pandruvada S, Royfman R, Shah TA, Sindhwani P, Dupree JM, Schon S, Avidor-Reiss T. Lack of trusted diagnostic tools for undetermined male infertility. J Assist Reprod Genet 2021; 38:265-276. [PMID: 33389378 PMCID: PMC7884538 DOI: 10.1007/s10815-020-02037-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022] Open
Abstract
Semen analysis is the cornerstone of evaluating male infertility, but it is imperfect and insufficient to diagnose male infertility. As a result, about 20% of infertile males have undetermined infertility, a term encompassing male infertility with an unknown underlying cause. Undetermined male infertility includes two categories: (i) idiopathic male infertility-infertile males with abnormal semen analyses with an unknown cause for that abnormality and (ii) unexplained male infertility-males with "normal" semen analyses who are unable to impregnate due to unknown causes. The treatment of males with undetermined infertility is limited due to a lack of understanding the frequency of general sperm defects (e.g., number, motility, shape, viability). Furthermore, there is a lack of trusted, quantitative, and predictive diagnostic tests that look inside the sperm to quantify defects such as DNA damage, RNA abnormalities, centriole dysfunction, or reactive oxygen species to discover the underlying cause. To better treat undetermined male infertility, further research is needed on the frequency of sperm defects and reliable diagnostic tools that assess intracellular sperm components must be developed. The purpose of this review is to uniquely create a paradigm of thought regarding categories of male infertility based on intracellular and extracellular features of semen and sperm, explore the prevalence of the various categories of male factor infertility, call attention to the lack of standardization and universal application of advanced sperm testing techniques beyond semen analysis, and clarify the limitations of standard semen analysis. We also call attention to the variability in definitions and consider the benefits towards undetermined male infertility if these gaps in research are filled.
Collapse
Affiliation(s)
- Swati Pandruvada
- Department of Biological Sciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43607 USA
| | - Rachel Royfman
- Department of Biological Sciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43607 USA
| | - Tariq A. Shah
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43607 USA
| | - Puneet Sindhwani
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43607 USA
| | - James M. Dupree
- Department of Urology and Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48019 USA
| | - Samantha Schon
- Division of Reproductive Endocrinology & Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Tomer Avidor-Reiss
- Department of Biological Sciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43607 USA
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43607 USA
| |
Collapse
|
17
|
Unnikrishnan V, Kastelic J, Thundathil J. Intracytoplasmic Sperm Injection in Cattle. Genes (Basel) 2021; 12:198. [PMID: 33572865 PMCID: PMC7911995 DOI: 10.3390/genes12020198] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 10/30/2022] Open
Abstract
Intracytoplasmic sperm injection (ICSI) involves the microinjection of sperm into a matured oocyte. Although this reproductive technology is successfully used in humans and many animal species, the efficiency of this procedure is low in the bovine species mainly due to failed oocyte activation following sperm microinjection. This review discusses various reasons for the low efficiency of ICSI in cattle, potential solutions, and future directions for research in this area, emphasizing the contributions of testis-specific isoforms of Na/K-ATPase (ATP1A4) and phospholipase C zeta (PLC ζ). Improving the efficiency of bovine ICSI would benefit the cattle breeding industries by effectively utilizing semen from elite sires at their earliest possible age.
Collapse
Affiliation(s)
| | | | - Jacob Thundathil
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N4N1, Canada; (V.U.); (J.K.)
| |
Collapse
|
18
|
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: 7.0] [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
|
19
|
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: 32] [Impact Index Per Article: 8.0] [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
|
20
|
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: 5.3] [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
|
21
|
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: 33] [Impact Index Per Article: 8.3] [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
|
22
|
Haghighat S, Tavalaee M, Kouhkan A, Zakeri Z, Noureddini M, Shahverdi AH, Nasr Esfahani MH. Reduction of truncated Kit Expression in Men with Abnormal Semen Parameters, Globozoospermia and History of Low or Fertilization Failure. CELL JOURNAL 2019; 21:314-321. [PMID: 31210438 PMCID: PMC6582429 DOI: 10.22074/cellj.2019.6112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/22/2018] [Indexed: 01/27/2023]
Abstract
Objective Phospholipase C zeta 1 (PLCζ) is one of the main sperm factor involved in oocyte activation and other
factors may assist this factor to induce successful fertilization. Microinjection of recombinant tr-kit, a truncated form of
c-kit receptor, into metaphase II-arrested mouse oocytes initiate egg activation. Considering the potential roles of tr-
KIT during spermiogenesis and fertilization, we aimed to assess expression of tr-KIT in sperm of men with normal and
abnormal parameters and also in infertile men with previous failed fertilization and globozoospermia.
Materials and Methods This experimental study was conducted from September 2015 to July 2016 on 30
normozoospermic and 20 abnormozoospermic samples for experiment one, and also was carried out on 10
globozoospermic men, 10 men with a history low or failed fertilization and 13 fertile men for experiment two. Semen
parameters and sperm DNA fragmentation were assessed according to WHO protocol, and TUNEL assay. Sperm tr-
KIT was evaluated by flow cytometry, immunostaining and western blot.
Results The results show that tr-KIT mainly was detected in post-acrosomal, equatorial and tail regions. Percentage
of tr-KIT-positive spermatozoa in abnormozoospermic men was significantly lower than normozoospermic men. Also
significant correlations were observed between sperm tr-KIT with sperm count (r=0.8, P<0.001), motility (r=0.31, P=0.03)
and abnormal morphology (r=-0.6, P<0.001). Expression of tr-KIT protein was significantly lower in infertile men with low/
failed fertilization and globozoospermia compared to fertile men. The significant correlation was also observed between
tr-KIT protein with fertilization rate (r=-0.46, P=0.04). In addition, significant correlations were observed between sperm
DNA fragmentation with fertilization rate (r=-0.56, P=0.019) and tr-KIT protein (r=-0.38, P=0.04).
Conclusion tr-KIT may play a direct or indirect role in fertilization. Therefore, to increase our insight regarding the role
of tr-KIT in fertilization further research is warranted.
Collapse
Affiliation(s)
- Somayeh Haghighat
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.,Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Marziyeh Tavalaee
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Azam Kouhkan
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Zahra Zakeri
- Department of Biology, Queens College and Graduate Center of the City University of New York, Flushing, NY, USA
| | - Mahdi Noureddini
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.Electronic Address:
| | - Abdol Hossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Medicine, ACECR, Tehran, Iran
| | - Mohammad Hossein Nasr Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.Electronic Address:.,Isfahan Fertility and Infertility Center, Isfahan, Iran
| |
Collapse
|
23
|
Fawzy M, Emad M, Mahran A, Sabry M, Fetih AN, Abdelghafar H, Rasheed S. Artificial oocyte activation with SrCl2 or calcimycin after ICSI improves clinical and embryological outcomes compared with ICSI alone: results of a randomized clinical trial. Hum Reprod 2019; 33:1636-1644. [PMID: 30099496 DOI: 10.1093/humrep/dey258] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Are pregnancy and birth rates affected by artificial oocyte activation (AOA) with SrCl2 or calcimycin after ICSI for couples with male-factor infertility linked to abnormal sperm morphology or for couples with previous ICSI cycles of unexplained low fertilization or inadequate fertilization associated with impaired oocyte morphology? SUMMARY ANSWER AOA with either SrCl2 or calcimycin can improve the rates of clinical pregnancy, ongoing pregnancy and live birth compared with ICSI alone, and the two agents have diverse effects for different subgroups of patients. WHAT IS KNOWN ALREADY ICSI is a successful treatment for infertility, but not in all individuals. AOA has potential to overcome inadequate fertilization in ICSI. Calcimycin and SrCl2 are candidate agents for AOA, but their effectiveness remains to be compared. STUDY DESIGN, SIZE, DURATION This study was a randomized, open-label, three-arm, parallel-group, double-centre, superiority trial conducted between April 2015 and January 2016. The study evaluated the effects of AOA with calcimycin or SrCl2 for clinical pregnancy rates after ICSI and included 343 couples divided into three groups. PARTICIPANTS/MATERIALS, SETTING, METHODS Couples were included if they had two previous ICSI cycles of no or low fertilization (0-30%) with unknown causes or impaired oocyte morphology. Male-factor infertility cycles (frozen-thawed sperm, surgically retrieved sperm or ejaculates contained <10 millions spermatozoa/ml) undergoing their first ICSI attempt were also included if they had 100% abnormal sperm morphology (including globozoospermia and tapered-head). Couples were randomized to undergo ICSI with SrCl2 AOA, ICSI with calcimycin AOA or ICSI alone, with clinical pregnancy as the primary endpoint. Effect sizes were summarized as absolute rate differences (ARDs) and odds ratios (ORs), with precision evaluated by 95% CIs. MAIN RESULTS AND THE ROLE OF CHANCE Both SrCl2 and calcimycin AOA improved clinical pregnancy rates compared to ICSI alone (49, 42 and 27%; ARD 22, 95% CI: 9-33; P = 0.0007 and ARD 16, 95% CI: 3-27; P = 0.014). SrCl2 and calcimycin AOA were also superior to ICSI alone on the rates of ongoing pregnancy (42, 36 and 23%; P = 0.0019 and P = 0.023) and live birth (40, 33 and 18%; P = 0.0002 and P = 0.012). Among couples with previous ICSI cycles of low fertilization, AOA with SrCl2 (but not with calcimycin) was superior to ICSI alone for rates of clinical pregnancy (ARD 35 percentage points (pp), P = 0.0007), ongoing pregnancy (ARD 27 pp, P = 0.009) and live birth (ARD 37 pp, P = 0.002). Among couples affected by male-factor infertility, AOA with calcimycin (but not with SrCl2) was superior to ICSI alone for rates of clinical pregnancy (ARD 22 pp, P = 0.006), ongoing pregnancy (ARD 19 pp, P = 0.013) and live birth (ARD 17 pp, P = 0.02). LIMITATIONS, REASONS FOR CAUTION This study was an open-label trial, and this design might have introduced bias, although randomization methods were used. The study did not include a longitudinal follow-up, so further evidence is required to demonstrate the safety of AOA. WIDER IMPLICATIONS OF THE FINDINGS The decision to use SrCl2 or calcimycin for AOA after ICSI may depend on whether the activation failure originates in the oocyte or the sperm. STUDY FUNDING/COMPETING INTEREST(S) The study received no funding and the authors have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER NCT02424214. TRIAL REGISTRATION DATE 22 April 2015. DATE OF FIRST PATIENT’S ENROLMENT 27 April 2015.
Collapse
Affiliation(s)
| | - Mai Emad
- IbnSina IVF Centre, IbnSina Hospital, Sohag, Egypt
| | - Ali Mahran
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Assiut University, AG, Egypt
| | - Mohamed Sabry
- Department of Obstetrics and Gynecology, Sohag University, Sohag, Egypt
| | - Ahmed N Fetih
- Department of Obstetrics and Gynecology, Faculty of Medicine, Assiut University, AG, Egypt
| | - Hazem Abdelghafar
- Department of Obstetrics and Gynecology, Sohag University, Sohag, Egypt
| | - Salah Rasheed
- Department of Obstetrics and Gynecology, Sohag University, Sohag, Egypt
| |
Collapse
|
24
|
Moreau J, Fargeon S, Gatimel N, Parinaud J, Léandri RD. Expression of phospholipase PLC Zeta in human spermatozoa: impact of cryopreservation. Andrology 2019; 7:315-318. [PMID: 30779311 DOI: 10.1111/andr.12593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/27/2018] [Accepted: 01/14/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cryopreservation is used for infertility treatment and for fertility preservation. The results of the use of frozen spermatozoa for ART (Assisted Reproductive Technology) are lower than those of fresh spermatozoa. The phospholipase C Zeta (PLCζ) protein is involved in oocyte activation. OBJECTIVES The aim of this study was to compare the percentage of spermatozoa expressing phospholipase C Zeta protein before and after a frozen-thawing cycle. MATERIALS AND METHODS Samples were provided after at least 2 days of sexual abstinence. A part of the fresh ejaculate (200 μL) was recovered for the study. Fifty microliters was necessary to carry out the technique before freezing. The remaining 150 μl was frozen according to a slow manual freezing technique. The samples were treated based on the procedure described by Yelumalai et al. (Fertil. Steril., 104, 2015, 561-568.e4) and Grasa et al. (Hum. Reprod. Oxf. Engl. 23, 2008, 2513-2522). RESULTS Freezing was associated with a decrease in the percentage of spermatozoa exhibiting PLCζ (44 ± 22% before vs 31 ± 19% after, p < 0.05). The percentage of spermatozoa exhibiting PLCζ at post-acrosomal position was significantly greater before freezing (8% vs 5%, p < 0.05). There was no significant difference for the percentage of spermatozoa exhibiting PLCζ at equatorial position (15% before freezing versus 12% after thawing, NS). DISCUSSION The results of the present study show that the presence of PLCζ on spermatozoa is decreased after freezing-thawing procedures. PLCζ is a soluble cytosolic protein (Nomikos et al., ), so it can be lost during cryopreservation. These membrane alterations are probably multifactorial. CONCLUSION Our results, in agreement with other studies, raise the hypothesis that cryopreservation reduces spermatic PLCζ expression.
Collapse
Affiliation(s)
- J Moreau
- Department of Reproductive Medicine, Toulouse University Hospital, Toulouse, France.,EA 3694 Human Fertility Research Group, Toulouse University Hospital, Toulouse, France
| | - S Fargeon
- EA 3694 Human Fertility Research Group, Toulouse University Hospital, Toulouse, France
| | - N Gatimel
- Department of Reproductive Medicine, Toulouse University Hospital, Toulouse, France.,EA 3694 Human Fertility Research Group, Toulouse University Hospital, Toulouse, France
| | - J Parinaud
- Department of Reproductive Medicine, Toulouse University Hospital, Toulouse, France.,EA 3694 Human Fertility Research Group, Toulouse University Hospital, Toulouse, France
| | - R D Léandri
- Department of Reproductive Medicine, Toulouse University Hospital, Toulouse, France.,EA 3694 Human Fertility Research Group, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
25
|
Parrington J, Arnoult C, Fissore RA. The eggstraordinary story of how life begins. Mol Reprod Dev 2018; 86:4-19. [PMID: 30411426 DOI: 10.1002/mrd.23083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 11/06/2018] [Indexed: 12/24/2022]
Abstract
More than 15 years have elapsed since the identification of phospholipase C ζ1 (PLCζ) from a genomic search for mouse testis/sperm-specific PLCs. This molecule was proposed to represent the sperm factor responsible for the initiation of calcium (Ca2+ ) oscillations required for egg activation and embryo development in mammals. Supporting evidence for this role emerged from studies documenting its expression in all mammals and other vertebrate species, the physiological Ca2+ rises induced by injection of its messenger RNA into mammalian and nonmammalian eggs, and the lack of expression in infertile males that fail intracytoplasmic sperm injection. In the last year, genetic animal models have added support to its role as the long sought-after sperm factor. In this review, we highlight the findings that demonstrated the role of Ca2+ as the universal signal of egg activation and the experimental buildup that culminated with the identification of PLCζ as the soluble sperm factor. We also discuss the structural-functional properties that make PLCζ especially suited to evoke oscillations in eggs. Lastly, we examine unresolved aspects of the function and regulation of PLCζ and whether or not it is the only sperm factor in mammalian sperm.
Collapse
Affiliation(s)
- John Parrington
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Christophe Arnoult
- Université Grenoble Alpes, Grenoble, France.,Institut pour l'Avancée des Biosciences (IAB), INSERM 1209, CNRS UMR 5309, La Tronche, France
| | - Rafael A Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts
| |
Collapse
|
26
|
Satouh Y, Ikawa M. New Insights into the Molecular Events of Mammalian Fertilization. Trends Biochem Sci 2018; 43:818-828. [PMID: 30170889 DOI: 10.1016/j.tibs.2018.08.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/30/2018] [Accepted: 08/04/2018] [Indexed: 12/20/2022]
Abstract
Currently, infertility affects ∼16% of couples worldwide. The causes are reported to involve both male and female factors, including fertilization failure between mature spermatozoa and eggs. However, the molecular mechanisms involved in each step of mammalian fertilization are yet to be fully elucidated. Although some of these steps can be rescued with assisted reproductive technologies, it is important to clarify the molecular mechanisms involved for the treatment and diagnosis of infertile couples. This review illustrates recent findings in mammalian fertilization, discovered by combining gene modification techniques with other new approaches, and aims to show how these findings will guide future research in mammalian fertilization.
Collapse
Affiliation(s)
- Yuhkoh Satouh
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan.
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan; The Institute of Medical Science, The University of Tokyo, Tokyo 1088639, Japan.
| |
Collapse
|
27
|
Goldbeter A. Dissipative structures in biological systems: bistability, oscillations, spatial patterns and waves. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2018; 376:rsta.2017.0376. [PMID: 29891498 PMCID: PMC6000149 DOI: 10.1098/rsta.2017.0376] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/26/2018] [Indexed: 05/05/2023]
Abstract
The goal of this review article is to assess how relevant is the concept of dissipative structure for understanding the dynamical bases of non-equilibrium self-organization in biological systems, and to see where it has been applied in the five decades since it was initially proposed by Ilya Prigogine. Dissipative structures can be classified into four types, which will be considered, in turn, and illustrated by biological examples: (i) multistability, in the form of bistability and tristability, which involve the coexistence of two or three stable steady states, or in the form of birhythmicity, which involves the coexistence between two stable rhythms; (ii) temporal dissipative structures in the form of sustained oscillations, illustrated by biological rhythms; (iii) spatial dissipative structures, known as Turing patterns; and (iv) spatio-temporal structures in the form of propagating waves. Rhythms occur with widely different periods at all levels of biological organization, from neural, cardiac and metabolic oscillations to circadian clocks and the cell cycle; they play key roles in physiology and in many disorders. New rhythms are being uncovered while artificial ones are produced by synthetic biology. Rhythms provide the richest source of examples of dissipative structures in biological systems. Bistability has been observed experimentally, but has primarily been investigated in theoretical models in an increasingly wide range of biological contexts, from the genetic to the cell and animal population levels, both in physiological conditions and in disease. Bistable transitions have been implicated in the progression between the different phases of the cell cycle and, more generally, in the process of cell fate specification in the developing embryo. Turing patterns are exemplified by the formation of some periodic structures in the course of development and by skin stripe patterns in animals. Spatio-temporal patterns in the form of propagating waves are observed within cells as well as in intercellular communication. This review illustrates how dissipative structures of all sorts abound in biological systems.This article is part of the theme issue 'Dissipative structures in matter out of equilibrium: from chemistry, photonics and biology (part 1)'.
Collapse
Affiliation(s)
- Albert Goldbeter
- Unité de Chronobiologie théorique, Service de Chimie physique et Biologie théorique, Faculté des Sciences, Université Libre de Bruxelles (ULB), Campus Plaine, CP 231, 1050 Brussels, Belgium
| |
Collapse
|
28
|
Satouh Y, Nozawa K, Yamagata K, Fujimoto T, Ikawa M. Viable offspring after imaging of Ca2+ oscillations and visualization of the cortical reaction in mouse eggs. Biol Reprod 2017; 96:563-575. [PMID: 28339615 DOI: 10.1093/biolre/iox002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/27/2017] [Indexed: 11/14/2022] Open
Abstract
– During mammalian fertilization, egg Ca 2+ oscillations are known to play pivotal roles in triggering downstream events such as resumption of the cell cycle and the establishment of blocks to polyspermy. However, viable offspring have not been obtained after monitoring Ca 2+ oscillations, and their spatiotemporal links to subsequent events are still to be examined. Therefore, the development of imaging methods to avoid phototoxic damage while labeling these events is required. Here, we examined the usefulness of genetically encoded Ca 2+ indicators for optical imaging (GECOs), in combination with spinning-disk confocal imaging. The Ca 2+ imaging of fertilized mouse eggs with GEM-, G-, or R-GECO recorded successful oscillations (8.19 ± 0.31, 7.56 ± 0.23, or 7.53 ± 0.27 spikes in the first 2 h, respectively), similar to those obtained with chemical indicators. Then, in vitro viability tests revealed that imaging with G- or R-GECO did not interfere with the rate of development to the blastocyst stage (61.8 or 70.0%, respectively, vs 75.0% in control). Furthermore, two-cell transfer to recipient female mice after imaging with G- or R-GECO resulted in a similar birthrate (53.3 or 52.0%, respectively) to that of controls (48.7%). Next, we assessed the quality of the cortical reaction (CR) in artificially activated or fertilized eggs using fluorescently labeled Lens culinaris agglutinin fluorescein isothiocyanate. Multicolor imaging demonstrated that the first few Ca 2+ spikes are sufficient for the completion of the CR and subsequent hardening of the zona pellucida in mouse eggs. These methods provide a framework for studying Ca 2+ dynamics in mammalian fertilization.
Collapse
Affiliation(s)
- Yuhkoh Satouh
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kaori Nozawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kazuo Yamagata
- Department of Genetic Engineering, Faculty of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama, Japan
| | - Takao Fujimoto
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
29
|
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: 3.6] [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
|
30
|
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: 3.0] [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
|
31
|
Abstract
Sustained oscillations abound in biological systems. They occur at all levels of biological organization over a wide range of periods, from a fraction of a second to years, and with a variety of underlying mechanisms. They control major physiological functions, and their dysfunction is associated with a variety of physiological disorders. The goal of this review is (i) to give an overview of the main rhythms observed at the cellular and supracellular levels, (ii) to briefly describe how the study of biological rhythms unfolded in the course of time, in parallel with studies on chemical oscillations, (iii) to present the major roles of biological rhythms in the control of physiological functions, and (iv) the pathologies associated with the alteration, disappearance, or spurious occurrence of biological rhythms. Two tables present the main examples of cellular and supracellular rhythms ordered according to their period, and their role in physiology and pathophysiology. Among the rhythms discussed are neural and cardiac rhythms, metabolic oscillations such as those occurring in glycolysis in yeast, intracellular Ca++ oscillations, cyclic AMP oscillations in Dictyostelium amoebae, the segmentation clock that controls somitogenesis, pulsatile hormone secretion, circadian rhythms which occur in all eukaryotes and some bacteria with a period close to 24 h, the oscillatory dynamics of the enzymatic network driving the cell cycle, and oscillations in transcription factors such as NF-ΚB and tumor suppressors such as p53. Ilya Prigogine's concept of dissipative structures applies to temporal oscillations and allows us to unify within a common framework the various rhythms observed at different levels of biological organization, regardless of their period and underlying mechanism.
Collapse
Affiliation(s)
- Albert Goldbeter
- Unité de Chronobiologie théorique, Service de Chimie physique et Biologie théorique, Faculté des Sciences, Université Libre de Bruxelles (ULB), Campus Plaine, CP 231, B-1050 Brussels, Belgium
| |
Collapse
|
32
|
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: 1.4] [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
|
33
|
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: 10.7] [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
|
34
|
Male infertility-linked point mutation reveals a vital binding role for the C2 domain of sperm PLCζ. Biochem J 2017; 474:1003-1016. [PMID: 28270562 DOI: 10.1042/bcj20161057] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/05/2017] [Accepted: 01/20/2017] [Indexed: 12/14/2022]
Abstract
Sperm-specific phospholipase C zeta (PLCζ) is widely considered to be the physiological stimulus that evokes intracellular calcium (Ca2+) oscillations that are essential for the initiation of egg activation during mammalian fertilisation. A recent genetic study reported a male infertility case that was directly associated with a point mutation in the PLCζ C2 domain, where an isoleucine residue had been substituted with a phenylalanine (I489F). Here, we have analysed the effect of this mutation on the in vivo Ca2+ oscillation-inducing activity and the in vitro biochemical properties of human PLCζ. Microinjection of cRNA or recombinant protein corresponding to PLCζI489F mutant at physiological concentrations completely failed to cause Ca2+ oscillations and trigger development. However, this infertile phenotype could be effectively rescued by microinjection of relatively high (non-physiological) amounts of recombinant mutant PLCζI489F protein, leading to Ca2+ oscillations and egg activation. Our in vitro biochemical analysis suggested that the PLCζI489F mutant displayed similar enzymatic properties, but dramatically reduced binding to PI(3)P and PI(5)P-containing liposomes compared with wild-type PLCζ. Our findings highlight the importance of PLCζ at fertilisation and the vital role of the C2 domain in PLCζ function, possibly due to its novel binding characteristics.
Collapse
|
35
|
Yamaguchi T, Ito M, Kuroda K, Takeda S, Tanaka A. The establishment of appropriate methods for egg-activation by human PLCZ1 RNA injection into human oocyte. Cell Calcium 2017; 65:22-30. [PMID: 28320563 DOI: 10.1016/j.ceca.2017.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/03/2017] [Accepted: 03/03/2017] [Indexed: 01/29/2023]
Abstract
Phospholipase C-zeta (PLCZ1), a strong candidate of egg-activating sperm factor, can induce Ca2+ oscillations and cause egg activation. For the application of PLCZ1 to clinical use, we examined the pattern of Ca2+ responses and developmental rate by comparing PLCZ1 RNA injection methods with the other current methods, such as cytosolic aspiration, electrical stimulation and ionomycin treatment in human oocytes. We found that the pattern of Ca2+ oscillations after PLCZ1 RNA injection exhibited similar characteristics to that after ICSI treatment. We also determined the optimal concentration of human PLCZ1 RNA to activate the human oocytes. Our findings suggest that human PLCZ1 RNA is a better therapeutic agent to rescue human oocytes from failed activation, leading to normal and efficient development.
Collapse
Affiliation(s)
- Takashi Yamaguchi
- Saint Mother Obstetrics and Gynecology Clinic, Institute for ART, Fukuoka 807-0825, Japan; Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Tokyo 113-0033, Japan
| | - Masahiko Ito
- Department of Virology and Parasitology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan.
| | - Keiji Kuroda
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Tokyo 113-0033, Japan
| | - Satoru Takeda
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, Tokyo 113-0033, Japan
| | - Atsushi Tanaka
- Saint Mother Obstetrics and Gynecology Clinic, Institute for ART, Fukuoka 807-0825, Japan
| |
Collapse
|
36
|
Cai X, Yu S, Mipam T, Yang F, Zhao W, Liu W, Cao S, Shen L, Zhao F, Sun L, Xu C, Wu S. Comparative analysis of testis transcriptomes associated with male infertility in cattleyak. Theriogenology 2017; 88:28-42. [DOI: 10.1016/j.theriogenology.2016.09.047] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 09/18/2016] [Accepted: 09/24/2016] [Indexed: 01/29/2023]
|
37
|
|
38
|
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: 13.0] [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
|
39
|
Brill JA, Yildirim S, Fabian L. Phosphoinositide signaling in sperm development. Semin Cell Dev Biol 2016; 59:2-9. [PMID: 27321976 DOI: 10.1016/j.semcdb.2016.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/15/2016] [Indexed: 01/15/2023]
Abstract
Phosphatidylinositol phosphates (PIPs)1 are membrane lipids with crucial roles during cell morphogenesis, including the establishment of cytoskeletal organization, membrane trafficking, cell polarity, cell-cycle control and signaling. Recent studies in mice (Mus musculus), fruit flies (Drosophila melanogaster) and other organisms have defined germ cell intrinsic requirements for these lipids and their regulatory enzymes in multiple aspects of sperm development. In particular, PIP levels are crucial in germline stem cell maintenance, spermatogonial proliferation and survival, spermatocyte cytokinesis, spermatid polarization, sperm tail formation, nuclear shaping, and production of mature, motile sperm. Here, we briefly review the stages of spermatogenesis and discuss the roles of PIPs and their regulatory enzymes in male germ cell development.
Collapse
Affiliation(s)
- Julie A Brill
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Sukriye Yildirim
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada.
| | - Lacramioara Fabian
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G OA4, Canada.
| |
Collapse
|
40
|
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: 4.3] [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
|
41
|
Denisenko O, Lucas ES, Sun C, Watkins AJ, Mar D, Bomsztyk K, Fleming TP. Regulation of ribosomal RNA expression across the lifespan is fine-tuned by maternal diet before implantation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:906-13. [PMID: 27060415 PMCID: PMC4914606 DOI: 10.1016/j.bbagrm.2016.04.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/18/2016] [Accepted: 04/04/2016] [Indexed: 01/17/2023]
Abstract
Cells and organisms respond to nutrient deprivation by decreasing global rates of transcription, translation and DNA replication. To what extent such changes can be reversed is largely unknown. We examined the effect of maternal dietary restriction on RNA synthesis in the offspring. Low protein diet fed either throughout gestation or for the preimplantation period alone reduced cellular RNA content across fetal somatic tissues during challenge and increased it beyond controls in fetal and adult tissues after challenge release. Changes in transcription of ribosomal RNA, the major component of cellular RNA, were responsible for this phenotype as evidenced by matching alterations in RNA polymerase I density and DNA methylation at ribosomal DNA loci. Cellular levels of the ribosomal transcription factor Rrn3 mirrored the rRNA expression pattern. In cell culture experiments, Rrn3 overexpression reduced rDNA methylation and increased rRNA expression; the converse occurred after inhibition of Rrn3 activity. These observations define novel mechanism where poor nutrition before implantation irreversibly alters basal rates of rRNA transcription thereafter in a process mediated by rDNA methylation and Rrn3 factor. Maternal malnutrition downregulates rDNA transcription in fetal tissues. Switch to normal diet permanently upregulates rDNA transcription compared to controls. These changes are mediated by DNA methylation and Pol I transcription factor Rrn3. This mechanism is activated before implantation.
Collapse
Affiliation(s)
- Oleg Denisenko
- Department of Medicine, University of Washington, 850 Republican St., Rm 242, Seattle, WA 98109, USA.
| | - Emma S Lucas
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - Congshan Sun
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - Adam J Watkins
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - Daniel Mar
- Department of Medicine, University of Washington, 850 Republican St., Rm 242, Seattle, WA 98109, USA
| | - Karol Bomsztyk
- Department of Medicine, University of Washington, 850 Republican St., Rm 242, Seattle, WA 98109, USA
| | - Tom P Fleming
- Centre for Biological Sciences, University of Southampton, Mailpoint 840, Level D Lab & Path Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| |
Collapse
|
42
|
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: 103] [Impact Index Per Article: 12.9] [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
|
43
|
Machaty Z. Signal transduction in mammalian oocytes during fertilization. Cell Tissue Res 2016; 363:169-183. [PMID: 26453398 PMCID: PMC4700098 DOI: 10.1007/s00441-015-2291-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/28/2015] [Indexed: 01/22/2023]
Abstract
Mammalian embryo development begins when the fertilizing sperm triggers a series of elevations in the oocyte's intracellular free Ca(2+) concentration. The elevations are the result of repeated release and re-uptake of Ca(2+) stored in the smooth endoplasmic reticulum. Ca(2+) release is primarily mediated by the phosphoinositide signaling system of the oocyte. The system is stimulated when the sperm causes the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG); IP3 then binds its receptor on the surface of the endoplasmic reticulum that induces Ca(2+) release. The manner in which the sperm generates IP3, the Ca(2+) mobilizing second messenger, has been the subject of extensive research for a long time. The sperm factor hypothesis has eventually gained general acceptance, according to which it is a molecule from the sperm that diffuses into the ooplasm and stimulates the phosphoinositide cascade. Much evidence now indicates that the sperm-derived factor is phospholipase C-zeta (PLCζ) that cleaves PIP2 and generates IP3, eventually leading to oocyte activation. A recent addition to the candidate sperm factor list is the post-acrosomal sheath WW domain-binding protein (PAWP), whose role at fertilization is currently under debate. Ca(2+) influx across the plasma membrane is also important as, in the absence of extracellular Ca(2+), the oscillations run down prematurely. In pig oocytes, the influx that sustains the oscillations seems to be regulated by the filling status of the stores, whereas in the mouse other mechanisms might be involved. This work summarizes the current understanding of Ca(2+) signaling in mammalian oocytes.
Collapse
Affiliation(s)
- Zoltan Machaty
- Department of Animal Sciences, Purdue University, 915 W. State Street, West Lafayette, IN, 47907, USA.
| |
Collapse
|
44
|
Fourteen babies born after round spermatid injection into human oocytes. Proc Natl Acad Sci U S A 2015; 112:14629-34. [PMID: 26575628 DOI: 10.1073/pnas.1517466112] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During the human in vitro fertilization procedure in the assisted reproductive technology, intracytoplasmic sperm injection is routinely used to inject a spermatozoon or a less mature elongating spermatid into the oocyte. In some infertile men, round spermatids (haploid male germ cells that have completed meiosis) are the most mature cells visible during testicular biopsy. The microsurgical injection of a round spermatid into an oocyte as a substitute is commonly referred to as round spermatid injection (ROSI). Currently, human ROSI is considered a very inefficient procedure and of no clinical value. Herein, we report the birth and development of 14 children born to 12 women following ROSI of 734 oocytes previously activated by an electric current. The round spermatids came from men who had been diagnosed as not having spermatozoa or elongated spermatids by andrologists at other hospitals after a first Micro-TESE. A key to our success was our ability to identify round spermatids accurately before oocyte injection. As of today, all children born after ROSI in our clinic are without any unusual physical, mental, or epigenetic problems. Thus, for men whose germ cells are unable to develop beyond the round spermatid stage, ROSI can, as a last resort, enable them to have their own genetic offspring.
Collapse
|
45
|
The Novel Functions of the PLC/PKC/PKD Signaling Axis in G Protein-Coupled Receptor-Mediated Chemotaxis of Neutrophils. J Immunol Res 2015; 2015:817604. [PMID: 26605346 PMCID: PMC4641950 DOI: 10.1155/2015/817604] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/05/2015] [Indexed: 12/16/2022] Open
Abstract
Chemotaxis, a directional cell migration guided by extracellular chemoattractant gradients, plays an essential role in the recruitment of neutrophils to sites of inflammation. Chemotaxis is mediated by the G protein-coupled receptor (GPCR) signaling pathway. Extracellular stimuli trigger activation of the PLC/PKC/PKD signaling axis, which controls several signaling pathways. Here, we concentrate on the novel functions of PLC/PKC/PKD signaling in GPCR-mediated chemotaxis of neutrophils.
Collapse
|
46
|
Nomikos M, Sanders JR, Parthimos D, Buntwal L, Calver BL, Stamatiadis P, Smith A, Clue M, Sideratou Z, Swann K, Lai FA. Essential Role of the EF-hand Domain in Targeting Sperm Phospholipase Cζ to Membrane Phosphatidylinositol 4,5-Bisphosphate (PIP2). J Biol Chem 2015; 290:29519-30. [PMID: 26429913 PMCID: PMC4705952 DOI: 10.1074/jbc.m115.658443] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Indexed: 11/25/2022] Open
Abstract
Sperm-specific phospholipase C-ζ (PLCζ) is widely considered to be the physiological stimulus that triggers intracellular Ca2+ oscillations and egg activation during mammalian fertilization. Although PLCζ is structurally similar to PLCδ1, it lacks a pleckstrin homology domain, and it remains unclear how PLCζ targets its phosphatidylinositol 4,5-bisphosphate (PIP2) membrane substrate. Recently, the PLCδ1 EF-hand domain was shown to bind to anionic phospholipids through a number of cationic residues, suggesting a potential mechanism for how PLCs might interact with their target membranes. Those critical cationic EF-hand residues in PLCδ1 are notably conserved in PLCζ. We investigated the potential role of these conserved cationic residues in PLCζ by generating a series of mutants that sequentially neutralized three positively charged residues (Lys-49, Lys-53, and Arg-57) within the mouse PLCζ EF-hand domain. Microinjection of the PLCζ EF-hand mutants into mouse eggs enabled their Ca2+ oscillation inducing activities to be compared with wild-type PLCζ. Furthermore, the mutant proteins were purified, and the in vitro PIP2 hydrolysis and binding properties were monitored. Our analysis suggests that PLCζ binds significantly to PIP2, but not to phosphatidic acid or phosphatidylserine, and that sequential reduction of the net positive charge within the first EF-hand domain of PLCζ significantly alters in vivo Ca2+ oscillation inducing activity and in vitro interaction with PIP2 without affecting its Ca2+ sensitivity. Our findings are consistent with theoretical predictions provided by a mathematical model that links oocyte Ca2+ frequency and the binding ability of different PLCζ mutants to PIP2. Moreover, a PLCζ mutant with mutations in the cationic residues within the first EF-hand domain and the XY linker region dramatically reduces the binding of PLCζ to PIP2, leading to complete abolishment of its Ca2+ oscillation inducing activity.
Collapse
Affiliation(s)
- Michail Nomikos
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Jessica R Sanders
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Dimitris Parthimos
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Luke Buntwal
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Brian L Calver
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Panagiotis Stamatiadis
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Adrian Smith
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Matthew Clue
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - Zili Sideratou
- the National Center for Scientific Research "Demokritos," 15310 Aghia Paraskevi, Greece
| | - Karl Swann
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| | - F Anthony Lai
- From the Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom and
| |
Collapse
|
47
|
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: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/13/2015] [Indexed: 12/11/2022] Open
|
48
|
Sanusi R, Yu Y, Nomikos M, Lai FA, Swann K. Rescue of failed oocyte activation after ICSI in a mouse model of male factor infertility by recombinant phospholipase Cζ. Mol Hum Reprod 2015; 21:783-91. [PMID: 26187950 PMCID: PMC4586348 DOI: 10.1093/molehr/gav042] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 07/13/2015] [Indexed: 12/17/2022] Open
Abstract
Artificial oocyte activation to overcome failed fertilization after intracytoplasmic sperm injection (ICSI) in human oocytes typically employs Ca(2+) ionophores to produce a single cytosolic Ca(2+) increase. In contrast, recombinant phospholipase Czeta (PLCζ) causes Ca(2+) oscillations indistinguishable from those occurring during fertilization, but remains untested for its efficacy in a scenario of ICSI fertilization failure. Here, we compare PLCζ with other activation stimuli in a mouse model of failed oocyte activation after ICSI, in which heat-treated sperm are injected into mouse oocytes. We show that increasing periods of 56 °C exposure of sperm produces a progressive loss of Ca(2+) oscillations after ICSI. The decrease in Ca(2+) oscillations produces a reduction in oocyte activation and embryo development to the blastocyst stage. We treated such oocytes that failed to activate after ICSI either with Ca(2+) ionophore, or with Sr(2+) media which causes Ca(2+) oscillations, or we injected them with recombinant human PLCζ. All these treatments rescued oocyte activation, although Sr(2+) and PLCζ gave the highest rates of development to blastocyst. When recombinant PLCζ was given to oocytes previously injected with control sperm, they developed normally to the blastocyst stage at rates similar to that after control ICSI. The data suggest that recombinant human PLCζ protein is an efficient means of rescuing oocyte activation after ICSI failure and that it can be effectively used even if the sperm already contains endogenous Ca(2+) releasing activity.
Collapse
Affiliation(s)
- Randa Sanusi
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Yuansong Yu
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK Present address: TopChoice Medical, TopChoice Building, 327 Tianmushan Road, Hangzhou City, Zhejiang Province, China
| | - Michail Nomikos
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - F Anthony Lai
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Karl Swann
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
49
|
Nomikos M, Sanders JR, Kashir J, Sanusi R, Buntwal L, Love D, Ashley P, Sanders D, Knaggs P, Bunkheila A, Swann K, Lai FA. Functional disparity between human PAWP and PLCζ in the generation of Ca2+ oscillations for oocyte activation. Mol Hum Reprod 2015; 21:702-10. [PMID: 26116451 DOI: 10.1093/molehr/gav034] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/19/2015] [Indexed: 11/14/2022] Open
Abstract
Mammalian oocyte activation is mediated by cytosolic calcium (Ca(2+)) oscillations initiated upon delivery of a putative 'sperm factor' by the fertilizing sperm. Previous studies suggest the identity of this sperm factor as the testis-specific phospholipase C-zeta (PLCζ). Recently, a post-acrosomal sheath WW domain-binding protein (PAWP) has been proposed as an alternative sperm factor candidate, following a report that human PAWP protein and cRNA elicited Ca(2+) oscillations in mouse and human oocytes. Those Ca(2+) oscillations were inhibited by a PAWP-derived peptide corresponding to a functional PPGY binding motif. Herein, using a series of human PAWP expression constructs, we demonstrate that both human PAWP protein and cRNA are, in our experiments, unable to elicit Ca(2+) release following microinjection into mouse oocytes. Parallel experiments performed with human PLCζ elicited the characteristic Ca(2+) oscillations present at mammalian fertilization, which produced oocyte activation and embryo development. Furthermore, sperm-induced Ca(2+) oscillations were not inhibited by the PAWP-derived PPGY peptide following in vitro fertilization or intracytoplasmic sperm injection. Thus, the functional disparity with PLCζ leads us to conclude that human PAWP is neither sufficient nor necessary for the Ca(2+) oscillations that initiate mammalian oocyte activation at fertilization.
Collapse
Affiliation(s)
- Michail Nomikos
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Jessica R Sanders
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Junaid Kashir
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - Randa Sanusi
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Luke Buntwal
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Daniel Love
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Peter Ashley
- Wales Fertility Institute, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - David Sanders
- Wales Fertility Institute, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Paul Knaggs
- Wales Fertility Institute, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Adnan Bunkheila
- Wales Fertility Institute, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Karl Swann
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - F Anthony Lai
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
50
|
Abstract
Egg activation is the first step of embryonic development and in mammals is triggered by a series of cytoplasmic calcium (Ca2+) oscillations. Sperm–egg fusion initiates these Ca2+ oscillations by introducing a sperm-specific protein factor into the egg cytoplasm. Substantial evidence indicates that this protein is a sperm-specific phospholipase C (PLC), termed PLC-zeta (PLCζ). PLCζ stimulates cytoplasmic Ca2+ oscillations matching those at fertilization triggering early embryonic development in several mammalian species. Structurally, PLCζ is comprised of four EF-hands, a C2 domain, and X and Y catalytic domains. PLCζ is an unusual PLC since it lacks a pleckstrin homology (PH) domain. It is also distinctive in that its X–Y linker is not involved in auto-inhibition of catalytic activity, but instead binds to phosphatidylinositol 4,5-bisphosphate (PIP2). Moreover, relative to other PLC isoforms, PLCζ possesses unique potency in stimulating Ca2+ oscillations in eggs, although it does not appear to bind to plasma membrane PIP2. In contrast, PLCζ appears to interact with intracellular vesicles in eggs that contain PIP2. I discuss the recent advances in our knowledge of the intriguing biochemical and physiological properties of sperm PLCζ and postulate potential roles for PLCζ in terms of clinical diagnosis and therapy for certain forms of male infertility.
Collapse
|