1
|
Brisard BM, Cashwell KD, Stewart SM, Harrison LM, Charles AC, Dennis CV, Henslee IR, Carrow EL, Belcher HA, Bhowmick D, Vos PW, Majka M, Bier M, Hart DM, Schmidt CA. Modeling diffusive search by non-adaptive sperm: Empirical and computational insights. PLoS Comput Biol 2025; 21:e1012865. [PMID: 40244975 PMCID: PMC12005489 DOI: 10.1371/journal.pcbi.1012865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/10/2025] [Indexed: 04/19/2025] Open
Abstract
During fertilization, mammalian sperm undergo a winnowing selection process that reduces the candidate pool of potential fertilizers from ~106-1011 cells to 101-102 cells (depending on the species). Classical sperm competition theory addresses the positive or 'stabilizing' selection acting on sperm phenotypes within populations of organisms but does not strictly address the developmental consequences of sperm traits among individual organisms that are under purifying selection during fertilization. It is the latter that is of utmost concern for improving assisted reproductive technologies (ART) because low-fitness sperm may be inadvertently used for fertilization during interventions that rely heavily on artificial sperm selection, such as intracytoplasmic sperm injection (ICSI). Importantly, some form of sperm selection is used in nearly all forms of ART (e.g., differential centrifugation, swim-up, or hyaluronan binding assays, etc.). To date, there is no unifying quantitative framework (i.e., theory of sperm selection) that synthesizes causal mechanisms of selection with observed natural variation in individual sperm traits. In this report, we reframe the physiological function of sperm as a collective diffusive search process and develop multi-scale computational models to explore the causal dynamics that constrain sperm fitness during fertilization. Several experimentally useful concepts are developed, including a probabilistic measure of sperm fitness as well as an information theoretic measure of the magnitude of sperm selection, each of which are assessed under systematic increases in microenvironmental selective pressure acting on sperm motility patterns.
Collapse
Affiliation(s)
- Benjamin M. Brisard
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Kylie D. Cashwell
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Stephanie M. Stewart
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Logan M. Harrison
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Aidan C. Charles
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Chelsea V. Dennis
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Ivie R. Henslee
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Ethan L. Carrow
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| | - Heather A. Belcher
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, North Carolina, United States of America
| | - Debajit Bhowmick
- Flow Cytometry Core Facility, Brody School of Medicine, East Carolina University, North Carolina, United States of America
| | - Paul W. Vos
- Department of Public Health, East Carolina University, Greenville, North Carolina, United States of America
| | - Maciej Majka
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Krakow, Poland
| | - Martin Bier
- Department of Physics, East Carolina University, Greenville, North Carolina, United States of America
| | - David M. Hart
- Department of Computer Science, East Carolina University, Greenville, North Carolina, United States of America
| | - Cameron A. Schmidt
- Department of Biology, East Carolina University, Greenville, North Carolina, United States of America
| |
Collapse
|
2
|
Xiong Y, Yi C, Zheng H, Ni Y, Xue Y, Li K. Protein palmitoylation is involved in regulating mouse sperm motility via the signals of calcium, protein tyrosine phosphorylation and reactive oxygen species. Biol Res 2025; 58:3. [PMID: 39810241 PMCID: PMC11734517 DOI: 10.1186/s40659-024-00580-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Protein palmitoylation, a critical posttranslational modification, plays an indispensable role in various cellular processes, including the regulation of protein stability, mediation of membrane fusion, facilitation of intracellular protein trafficking, and participation in cellular signaling pathways. It is also implicated in the pathogenesis of diseases, such as cancer, neurological disorders, inflammation, metabolic disorders, infections, and neurodegenerative diseases. However, its regulatory effects on sperm physiology, particularly motility, remain unclear. This study aimed to elucidate the mechanism by which protein palmitoylation governs sperm motility. METHODS Protein palmitoylation in situ in mouse sperm was observed using innovative click chemistry. Sperm motility and motion parameters were evaluated using a computer-assisted sperm analyzer (CASA) after treatment with 2-bromopalmitic acid (2BP), a specific inhibitor of protein palmitoylation. Protein palmitoylation levels were confirmed by the acyl-biotin exchange (ABE) method. The interplay between protein palmitoylation, protein tyrosine phosphorylation, and intracellular calcium was investigated using Western blotting, ABE method, and fluorescent probes. The regulation of reactive oxygen species was also examined using fluorescent probes. RESULTS Localized patterns and dynamics of protein palmitoylation in distinct sperm regions were revealed, including the midpiece, post-acrosomal region, acrosome, and head. Alterations in protein palmitoylation in sperm were observed under in vitro physiological conditions. Treatment with 2BP significantly affected sperm motility and motion parameters. The study revealed interactions between protein palmitoylation, including heat shock protein 90, and protein kinase A/protein kinase C-associated protein tyrosine phosphorylation and intracellular calcium. Additionally, protein palmitoylation was found to be involved in reactive oxygen species regulation. CONCLUSIONS Protein palmitoylation regulates sperm motility through calcium signaling, protein tyrosine phosphorylation, and reactive oxygen species. This study revealed the characteristics of protein palmitoylation in sperm and its role in regulating sperm motility, thereby providing novel insights into the causes of asthenozoospermia associated with sperm motility in humans.
Collapse
Affiliation(s)
- Yuping Xiong
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenchen Yi
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Haixia Zheng
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ya Ni
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yamei Xue
- Department of Obstetrics and Gynecology, Assisted Reproduction Unit, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Kun Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Martínez-Pastor F, Ramón M. Bioinformatics on Sperm Subpopulations Using Computer-Assisted Sperm Analysis (CASA). Methods Mol Biol 2025; 2897:235-247. [PMID: 40202640 DOI: 10.1007/978-1-0716-4406-5_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Computer-aided sperm analysis of motility (CASA-mot) and morphology (CASA-morph) enables the quick acquisition of many parameters on individual spermatozoa. This results in massive datasets apt for exploration using multiparametric tools. Data clustering has been used for decades to identify subpopulations of spermatozoa sharing similar motility patterns or morphological features. There are two main strategies considering the lack of or availability of prior information: unsupervised and supervised clustering. Then, the researcher has a wealth of algorithms for processing the data in both approaches, but one has to be aware that not all of them could be appropriate for dealing with the peculiarities of sperm data. Here, we describe in detail two approaches previously published: the two-step procedure as unsupervised and the support vector machines (SVM) as supervised. Both approaches are widely available in different statistical software, but in the interest of openness in research, specific details are provided for the R statistical environment (open source and non-proprietary platform and packages). This chapter offers strengths, caveats, and suggestions for alternative algorithms.
Collapse
Affiliation(s)
- Felipe Martínez-Pastor
- Institute of Animal Health and Cattle Development (INDEGSAL) and Department of Molecular Biology (Cell Biology), Universidad de León, León, Spain
| | - Manuel Ramón
- CERSYRA-IRIAF, Valdepeñas, Ciudad Real, Spain.
- Mejora Genética Animal, INIA-CSIC, Madrid, Spain.
| |
Collapse
|
4
|
Brisard BM, Cashwell KD, Stewart SM, Harrison LM, Charles AC, Dennis CV, Henslee IR, Carrow EL, Belcher HA, Bhowmick D, Vos PW, Majka M, Bier M, Hart DM, Schmidt CA. Modeling Diffusive Search by Non-Adaptive Sperm: Empirical and Computational Insights. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599386. [PMID: 38948799 PMCID: PMC11212867 DOI: 10.1101/2024.06.17.599386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
During fertilization, mammalian sperm undergo a winnowing selection process that reduces the candidate pool of potential fertilizers from ~106-1011 cells to 101-102 cells (depending on the species). Classical sperm competition theory addresses the positive or 'stabilizing' selection acting on sperm phenotypes within populations of organisms but does not strictly address the developmental consequences of sperm traits among individual organisms that are under purifying selection during fertilization. It is the latter that is of utmost concern for improving assisted reproductive technologies (ART) because 'low fitness' sperm may be inadvertently used for fertilization during interventions that rely heavily on artificial sperm selection, such as intracytoplasmic sperm injection (ICSI). Importantly, some form of sperm selection is used in nearly all forms of ART (e.g., differential centrifugation, swim-up, or hyaluronan binding assays, etc.). To date, there is no unifying quantitative framework (i.e., theory of sperm selection) that synthesizes causal mechanisms of selection with observed natural variation in individual sperm traits. In this report, we reframe the physiological function of sperm as a collective diffusive search process and develop multi-scale computational models to explore the causal dynamics that constrain sperm 'fitness' during fertilization. Several experimentally useful concepts are developed, including a probabilistic measure of sperm 'fitness' as well as an information theoretic measure of the magnitude of sperm selection, each of which are assessed under systematic increases in microenvironmental selective pressure acting on sperm motility patterns.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ivie R. Henslee
- Department of Biology, East Carolina University, Greenville NC
| | - Ethan L. Carrow
- Department of Biology, East Carolina University, Greenville NC
| | - Heather A. Belcher
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University
| | - Debajit Bhowmick
- Flow Cytometry Core Facility, Brody School of Medicine, East Carolina University
| | - Paul W. Vos
- Department of Public Health, East Carolina University, Greenville NC
| | - Maciej Majka
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Krakow, Poland
| | - Martin Bier
- Department of Physics, East Carolina University, Greenville NC
| | - David M. Hart
- Department of Computer Science, East Carolina University, Greenville, NC
| | | |
Collapse
|
5
|
Cecchini K, Ajaykumar N, Bagci A, Zamore PD, Gainetdinov I. Mouse Pachytene piRNAs Cleave Hundreds of Transcripts, But Alter the Steady-State Abundance of Only a Minority of Targets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.02.621675. [PMID: 39554027 PMCID: PMC11566022 DOI: 10.1101/2024.11.02.621675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
In animals, 18-35-nt piRNAs guide PIWI proteins to regulate complementary RNAs. During male meiosis, mammals produce an exceptionally abundant class of piRNAs called pachytene piRNAs. Pachytene piRNAs are required for spermatogenesis and have been proposed to control gene expression by various mechanisms. Here, we show that pachytene piRNAs regulate targets predominantly, if not exclusively, by endonucleolytic cleavage. Remarkably, pachytene piRNAs slice hundreds of RNAs, yet a change in steady-state level is detectable for a small fraction of transcripts. Our data suggest that cleavage of the few targets whose abundance is reduced significantly by piRNAs is essential for male fertility. Other pachytene piRNA targets are enriched for highly transcribed genes, which may explain why piRNA cleavage is often inconsequential for the steady-state abundance of targets. We propose that the retention of pachytene piRNAs throughout mammalian evolution is driven by the selective advantage conferred by a tiny minority of piRNAs.
Collapse
Affiliation(s)
- Katharine Cecchini
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Ayca Bagci
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Phillip D Zamore
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|
6
|
Castro M, Aguila L, Arias ME, Felmer R. Production of bovine embryos by piezo-ICSI using capacitated spermatozoa selected by fluorescence-activated cell sorting (FACS-piezo-ICSI). Anim Reprod Sci 2024; 268:107560. [PMID: 39029370 DOI: 10.1016/j.anireprosci.2024.107560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/11/2024] [Accepted: 07/07/2024] [Indexed: 07/21/2024]
Abstract
Intracytoplasmic sperm injection (ICSI) remains inefficient in cattle. One reason could lie in the injection of oocytes with sperm that have not undergone molecular changes associated with in vivo capacitation and fertilizing ability. This study aimed to enhance the efficiency of bovine intracytoplasmic sperm injection (piezo-ICSI) by employing fluorescent-activated cell sorting (FACS) to select the sperm population before injection based on capacitation markers. First, we evaluated the effects of incubating thawed sperm for 2 hours with different capacitating inductors: heparin, methyl-beta-cyclodextrin (MβCD), and dibutyryl cyclic AMP (dbcAMP), alone or in combinations in a basal capacitating (C) medium (Sp-TALP). Sperm capacitation and quality markers were evaluated by flow cytometry, revealing heparin as the most effective inducer of sperm capacitation changes. It, therefore, this treatment was chosen as the sperm pretreatment for FACS-piezo-ICSI. Two cell populations showing high capacitating levels (Heparin-HCL) and low capacitating levels (Heparin-LCL) of the markers associated with sperm capacitation i(Ca2+) levels and acrosome integrity were selected by FACS and used for sperm injection. Pronuclear formation was significantly higher when ICSI was performed with Heparin-HCL sperm than with Heparin-LCL and the control group (Heparin unsorted) groups (50 %, 10 %, and 20 %, respectively). Furthermore, injecting Heparin-HCL sperm resulted in a higher blastocyst rate (22.5 %) than Heparin-LCL (10 %) and the control group (15.2 %). In conclusion, heparin treatment effectively induced changes associated with sperm capacitation. The combination of Heparin-HCL treatment and FACS enabled precise selection of capacitated sperm before ICSI, enhancing the efficiency of this technology in the bovine species.
Collapse
Affiliation(s)
- Macarena Castro
- Laboratory of Reproduction, Center of Reproductive Biotechnology (CEBIOR-BIOREN), Faculty of Medicine, Universidad de la Frontera, Temuco, Chile; Master of Science Program specializing in Biology of Reproduction, Universidad de La Frontera, Temuco, Chile
| | - Luis Aguila
- Laboratory of Reproduction, Center of Reproductive Biotechnology (CEBIOR-BIOREN), Faculty of Medicine, Universidad de la Frontera, Temuco, Chile
| | - María Elena Arias
- Laboratory of Reproduction, Center of Reproductive Biotechnology (CEBIOR-BIOREN), Faculty of Medicine, Universidad de la Frontera, Temuco, Chile; Department of Agricultural Production, Faculty of Agriculture and Environmental Sciences, Universidad de La Frontera, Temuco, Chile
| | - Ricardo Felmer
- Laboratory of Reproduction, Center of Reproductive Biotechnology (CEBIOR-BIOREN), Faculty of Medicine, Universidad de la Frontera, Temuco, Chile; Department of Agricultural Sciences and Natural Resources, Faculty of Agricultural and Environmental Sciences, Universidad de La Frontera, Temuco, Chile.
| |
Collapse
|
7
|
Karimi A, Kohpeyma F, Asadi E, Ziyaee M, Karimi S. Protective efficacy of Nerium oleander extract on spermatogenesis in streptozotocin-induced diabetic rats. ZYGOTE 2024; 32:139-148. [PMID: 38284273 DOI: 10.1017/s0967199423000643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Men with diabetes frequently experience spermatogenic dysfunction, which is the most significant sign that diabetes has harmed their ability to reproduce. The effect of various doses of the hydro-alcoholic extract of Nerium oleander leaves on the pituitary-gonadal axis, sperm motility and number, antioxidant system, changes in testicular tissue structure, and spermatogenesis in healthy and diabetic rats has been examined in the current study. Eighty male rats that had been streptozotocin-induced diabetic and healthy were divided into eight groups: (1) control, (2) Nerium (50 mg/kg), (3) Nerium (100 mg/kg), (4) Nerium (200 mg/kg), (5) DM (6) DM+Nerium (50 mg/kg), (7) DM+Nerium (100 mg/kg) and (8) DM+Nerium (200 mg/kg) and were administered orally for 48 days consecutive. Following the studies, analysis of the testicular tissues' antioxidant capacity as well as sperm parameters, Johnsen's scoring and morphometric evaluation, histology, biochemical and stereology studies were performed.The outcomes showed that Nerium 50 and 100 mg/kg considerably enhanced the testicular morphology, sperm parameters, and reproductive organs to varying degrees in diabetic rats. After Nerium 50 mg/kg administration, glutathione peroxidase (GPX) and catalase (CAT) levels in the testicular tissue were increased whereas malondialdehyde (MDA) levels were markedly decreased. Nerium may help protect against diabetic-induced spermatogenic dysfunction in male rats by enhancing the activities of antioxidant enzymes in lower dosages.
Collapse
Affiliation(s)
- Afrooz Karimi
- Department of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Kohpeyma
- Endocrine and Metabolism Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Ebrahim Asadi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | | | - Samaneh Karimi
- Department of Anatomical Sciences, School of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| |
Collapse
|
8
|
Peña FJ, Martín-Cano FE, Becerro-Rey L, Ortega-Ferrusola C, Gaitskell-Phillips G, da Silva-Álvarez E, Gil MC. The future of equine semen analysis. Reprod Fertil Dev 2024; 36:RD23212. [PMID: 38467450 DOI: 10.1071/rd23212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
We are currently experiencing a period of rapid advancement in various areas of science and technology. The integration of high throughput 'omics' techniques with advanced biostatistics, and the help of artificial intelligence, is significantly impacting our understanding of sperm biology. These advances will have an appreciable impact on the practice of reproductive medicine in horses. This article provides a brief overview of recent advances in the field of spermatology and how they are changing assessment of sperm quality. This article is written from the authors' perspective, using the stallion as a model. We aim to portray a brief overview of the changes occurring in the assessment of sperm motility and kinematics, advances in flow cytometry, implementation of 'omics' technologies, and the use of artificial intelligence/self-learning in data analysis. We also briefly discuss how some of the advances can be readily available to the practitioner, through the implementation of 'on-farm' devices and telemedicine.
Collapse
Affiliation(s)
- Fernando J Peña
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Francisco Eduardo Martín-Cano
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Laura Becerro-Rey
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Cristina Ortega-Ferrusola
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Gemma Gaitskell-Phillips
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - Eva da Silva-Álvarez
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| | - María Cruz Gil
- Laboratory of Equine Reproduction and Equine Spermatology, Veterinary Teaching Hospital, University of Extremadura, Cáceres, Spain
| |
Collapse
|
9
|
Yang M, Diaz F, Krause ART, Lei Y, Liu WS. Synergistic enhancement of the mouse Pramex1 and Pramel1 in repressing retinoic acid (RA) signaling during gametogenesis. Cell Biosci 2024; 14:28. [PMID: 38395975 PMCID: PMC10893636 DOI: 10.1186/s13578-024-01212-w] [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: 12/07/2023] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND PRAME constitutes one of the largest multi-copy gene families in Eutherians, encoding cancer-testis antigens (CTAs) with leucine-rich repeats (LRR) domains, highly expressed in cancer cells and gametogenic germ cells. This study aims to elucidate genetic interactions between two members, Pramex1 and Pramel1, in the mouse Prame family during gametogenesis using a gene knockout approach. RESULT Single-gene knockout (sKO) of either Pramex1 or Pramel1 resulted in approximately 7% of abnormal seminiferous tubules, characterized by a Sertoli-cell only (SCO) phenotype, impacting sperm count and fecundity significantly. Remarkably, sKO female mice displayed normal reproductive functions. In contrast, Pramex1/Pramel1 double knockout (dKO) mice exhibited reduced fecundity in both sexes. In dKO females, ovarian primary follicle count decreased by 50% compared to sKO and WT mice, correlating with a 50% fecundity decrease. This suggested compensatory roles during oogenesis in Pramex1 or Pramel1 sKO females. Conversely, dKO males showed an 18% frequency of SCO tubules, increased apoptotic germ cells, and decreased undifferentiated spermatogonia compared to sKO and WT testes. Western blot analysis with PRAMEX1- or PRAMEL1-specific antibodies on sKO testes revealed compensatory upregulation of each protein (30-50%) in response to the other gene's deletion. Double KO males exhibited more severe defects in sperm count and litter size, surpassing Pramex1 and Pramel1 sKO accumulative effects, indicating a synergistic enhancement interaction during spermatogenesis. Additional experiments administering trans-retinoic acid (RA) and its inhibitor (WIN18,446) in sKO, dKO, and WT mice suggested that PRAMEX1 and PRAMEL1 synergistically repress the RA signaling pathway during spermatogenesis. CONCLUSION Data from sKO and dKO mice unveil a synergistic interaction via the RA signaling pathway between Pramex1 and Pramel1 genes during gametogenesis. This discovery sets the stage for investigating interactions among other members within the Prame family, advancing our understanding of multi-copy gene families involved in germ cell formation and function.
Collapse
Affiliation(s)
- Mingyao Yang
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Francisco Diaz
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Ana Rita T Krause
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA
| | - Yuguo Lei
- Department of Biomedical Engineering, College of Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Wan-Sheng Liu
- Department of Animal Science, Center for Reproductive Biology and Health (CRBH), College of Agricultural Sciences, The Pennsylvania State University, 311 AVBS Building, University Park, PA, 16802, USA.
| |
Collapse
|
10
|
Schmidt CA, Hale BJ, Bhowmick D, Miller WJ, Neufer PD, Geyer CB. Pyruvate modulation of redox potential controls mouse sperm motility. Dev Cell 2024; 59:79-90.e6. [PMID: 38101411 PMCID: PMC10872278 DOI: 10.1016/j.devcel.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 06/21/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023]
Abstract
Sperm gain fertilization competence in the female reproductive tract through a series of biochemical changes and a requisite switch from linear progressive to hyperactive motility. Despite being essential for fertilization, regulation of sperm energy transduction is poorly understood. This knowledge gap confounds interpretation of interspecies variation and limits progress in optimizing sperm selection for assisted reproduction. Here, we developed a model of mouse sperm bioenergetics using metabolic phenotyping data, quantitative microscopy, and spectral flow cytometry. The results define a mechanism of motility regulation by microenvironmental pyruvate. Rather than being consumed as a mitochondrial fuel source, pyruvate stimulates hyperactivation by repressing lactate oxidation and activating glycolysis in the flagellum through provision of nicotinamide adenine dinucleotide (NAD)+. These findings provide evidence that the transitions in motility requisite for sperm competence are governed by changes in the metabolic microenvironment, highlighting the unexplored potential of using catabolite combination to optimize sperm selection for fertilization.
Collapse
Affiliation(s)
- Cameron A Schmidt
- Department of Biology at East Carolina University, Greenville, NC 27858, USA; East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NC 27834, USA.
| | - Benjamin J Hale
- East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NC 27834, USA; Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Debajit Bhowmick
- Flow Cytometry Core Facility, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - William J Miller
- Department of Engineering, East Carolina University, Greenville, NC 27834, USA
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NC 27834, USA; Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Christopher B Geyer
- East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NC 27834, USA; Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
11
|
Ghayda RA, Cannarella R, Calogero AE, Shah R, Rambhatla A, Zohdy W, Kavoussi P, Avidor-Reiss T, Boitrelle F, Mostafa T, Saleh R, Toprak T, Birowo P, Salvio G, Calik G, Kuroda S, Kaiyal RS, Ziouziou I, Crafa A, Phuoc NHV, Russo GI, Durairajanayagam D, Al-Hashimi M, Hamoda TAAAM, Pinggera GM, Adriansjah R, Maldonado Rosas I, Arafa M, Chung E, Atmoko W, Rocco L, Lin H, Huyghe E, Kothari P, Solorzano Vazquez JF, Dimitriadis F, Garrido N, Homa S, Falcone M, Sabbaghian M, Kandil H, Ko E, Martinez M, Nguyen Q, Harraz AM, Serefoglu EC, Karthikeyan VS, Tien DMB, Jindal S, Micic S, Bellavia M, Alali H, Gherabi N, Lewis S, Park HJ, Simopoulou M, Sallam H, Ramirez L, Colpi G, Agarwal A. Artificial Intelligence in Andrology: From Semen Analysis to Image Diagnostics. World J Mens Health 2024; 42:39-61. [PMID: 37382282 PMCID: PMC10782130 DOI: 10.5534/wjmh.230050] [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: 03/02/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 06/30/2023] Open
Abstract
Artificial intelligence (AI) in medicine has gained a lot of momentum in the last decades and has been applied to various fields of medicine. Advances in computer science, medical informatics, robotics, and the need for personalized medicine have facilitated the role of AI in modern healthcare. Similarly, as in other fields, AI applications, such as machine learning, artificial neural networks, and deep learning, have shown great potential in andrology and reproductive medicine. AI-based tools are poised to become valuable assets with abilities to support and aid in diagnosing and treating male infertility, and in improving the accuracy of patient care. These automated, AI-based predictions may offer consistency and efficiency in terms of time and cost in infertility research and clinical management. In andrology and reproductive medicine, AI has been used for objective sperm, oocyte, and embryo selection, prediction of surgical outcomes, cost-effective assessment, development of robotic surgery, and clinical decision-making systems. In the future, better integration and implementation of AI into medicine will undoubtedly lead to pioneering evidence-based breakthroughs and the reshaping of andrology and reproductive medicine.
Collapse
Affiliation(s)
- Ramy Abou Ghayda
- Urology Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rupin Shah
- Department of Urology, Lilavati Hospital and Research Centre, Mumbai, India
| | - Amarnath Rambhatla
- Department of Urology, Henry Ford Health System, Vattikuti Urology Institute, Detroit, MI, USA
| | - Wael Zohdy
- Andrology and STDs, Cairo University, Cairo, Egypt
| | - Parviz Kavoussi
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tomer Avidor-Reiss
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Florence Boitrelle
- Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment, and Development, Paris Saclay University, UVSQ, INRAE, BREED, Paris, France
| | - Taymour Mostafa
- Andrology, Sexology & STIs Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ramadan Saleh
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Tuncay Toprak
- Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ponco Birowo
- Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Gianmaria Salvio
- Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy
| | - Gokhan Calik
- Department of Urology, Istanbul Medipol University, Istanbul, Turkey
| | - Shinnosuke Kuroda
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Urology, Reproduction Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Raneen Sawaid Kaiyal
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Imad Ziouziou
- Department of Urology, College of Medicine and Pharmacy, Ibn Zohr University, Agadir, Morocco
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nguyen Ho Vinh Phuoc
- Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
- Department of Urology and Andrology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | | | - Damayanthi Durairajanayagam
- Department of Physiology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Manaf Al-Hashimi
- Department of Urology, Burjeel Hospital, Abu Dhabi, United Arab Emirates (UAE)
- Khalifa University, College of Medicine and Health Science, Abu Dhabi, United Arab Emirates (UAE)
| | - Taha Abo-Almagd Abdel-Meguid Hamoda
- Department of Urology, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Urology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | | | - Ricky Adriansjah
- Department of Urology, Hasan Sadikin General Hospital, Universitas Padjadjaran, Banding, Indonesia
| | | | - Mohamed Arafa
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, Weill Cornell Medical-Qatar, Doha, Qatar
| | - Eric Chung
- Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane QLD, Australia
| | - Widi Atmoko
- Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Lucia Rocco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
| | - Eric Huyghe
- Department of Urology and Andrology, University Hospital of Toulouse, Toulouse, France
| | - Priyank Kothari
- Department of Urology, B.Y.L. Nair Charitable Hospital, Topiwala National Medical College, Mumbai, India
| | | | - Fotios Dimitriadis
- Department of Urology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nicolas Garrido
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Sheryl Homa
- Department of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Marco Falcone
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Marjan Sabbaghian
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | - Edmund Ko
- Department of Urology, Loma Linda University Health, Loma Linda, CA, USA
| | - Marlon Martinez
- Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines
| | - Quang Nguyen
- Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines
- Center for Andrology and Sexual Medicine, Viet Duc University Hospital, Hanoi, Vietnam
- Department of Urology, Andrology and Sexual Medicine, University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Ahmed M. Harraz
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
- Department of Surgery, Urology Unit, Farwaniya Hospital, Farwaniya, Kuwait
- Department of Urology, Sabah Al Ahmad Urology Center, Kuwait City, Kuwait
| | - Ege Can Serefoglu
- Department of Urology, Biruni University School of Medicine, Istanbul, Turkey
| | | | - Dung Mai Ba Tien
- Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
| | - Sunil Jindal
- Department of Andrology and Reproductive Medicine, Jindal Hospital, Meerut, India
| | - Sava Micic
- Department of Andrology, Uromedica Polyclinic, Belgrade, Serbia
| | - Marina Bellavia
- Andrology and IVF Center, Next Fertility Procrea, Lugano, Switzerland
| | - Hamed Alali
- King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Nazim Gherabi
- Andrology Committee of the Algerian Association of Urology, Algiers, Algeria
| | - Sheena Lewis
- Examen Lab Ltd., Northern Ireland, United Kingdom
| | - Hyun Jun Park
- Department of Urology, Pusan National University School of Medicine, Busan, Korea
- Medical Research Institute of Pusan National University Hospital, Busan, Korea
| | - Mara Simopoulou
- Department of Experimental Physiology, School of Health Sciences, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Hassan Sallam
- Alexandria University Faculty of Medicine, Alexandria, Egypt
| | - Liliana Ramirez
- IVF Laboratory, CITMER Reproductive Medicine, Mexico City, Mexico
| | - Giovanni Colpi
- Andrology and IVF Center, Next Fertility Procrea, Lugano, Switzerland
| | - Ashok Agarwal
- Global Andrology Forum, Moreland Hills, OH, USA
- Cleveland Clinic, Cleveland, OH, USA
| | | |
Collapse
|
12
|
Mahmoud M, Abd-Allah SM, Abdel-Halim BR, Khalil AAY. Ameliorative effect of chitosan nanoparticles in capacitation media on post-thawing in vitro fertilizing ability of bovine spermatozoa. Reprod Domest Anim 2023; 58:1428-1438. [PMID: 37635322 DOI: 10.1111/rda.14458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/29/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023]
Abstract
This study aimed to investigate the effect of supplementation of chitosan nanoparticles (CSNPs) on the capacitation of bovine spermatozoa during the in vitro fertilization process. Hyperactivated motility (HAM) and acrosome reaction (AR) of sperm cells as well as in vitro fertilization and cleavage rates are the main parameters used to estimate the effect of CSNPs on bovine spermatozoa's fertilizing ability. In this study, three different concentrations of CSNPs (10, 20 and 100 μg/mL) were prepared and characterized. Motile spermatozoa were separated from frozen-thawed semen by a swim-up technique and capacitated in Sperm-TALP medium supplemented with heparin only without CSNPs treatment (positive control), heparin + 10 μg/mL CSNPs, heparin + 20 μg/mL CSNPs, heparin + 100 μg/mL CSNPs and the last one served as a negative control tube which supplemented with 10 μg/mL CSNPs without adding heparin. Sperm cells were incubated for 90 min at 39°C in a 5% CO2 incubator and evaluated every 30 min at intervals. Cumulus oophorus complex (COCs) were matured in a 5% CO2 incubator at 39°C and inseminated in vitro with frozen-thawed bull sperm of the above concentrations. The inseminated oocytes were incubated at 39°C in a 5% CO2 incubator for 24 h and then examined for evidence of fertilization. The results of this investigation showed that HAM and AR were best affected by CSNPs at a concentration of 20 μg/mL during an incubation time of 60 min. As time went on, the overall proportion of spermatozoa with progressive motility (PM) decreased across all groups, and a substantially lower value was found at the dose mentioned above. Additionally, the impact of sperm treated with CSNPs on fertilization rate was assessed. The outcomes demonstrated that in comparison to the other concentrations (10 and 100 μg/mL), the positive control and the negative control, the proportion of fertilized oocytes was significantly higher in the CSNPs concentration (20 μg/mL). In conclusion, it could be inferred from this investigation that CSNPs support sperm functions during IVF and can be used for biomedical interventions in bovine spermatozoa. Additionally, a high IVF rate was achieved by using sperm treated with CSNPs as CSNPs enhance sperm capacitation and acrosome reaction.
Collapse
Affiliation(s)
- Mona Mahmoud
- Department of Theriogenology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Saber M Abd-Allah
- Department of Theriogenology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Bakar R Abdel-Halim
- Department of Theriogenology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Abdeltawab A Y Khalil
- Department of Theriogenology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
13
|
Rodríguez-Martínez EA, Rivas CU, Ayala ME, Blanco-Rodríguez R, Juarez N, Hernandez-Vargas EA, Aragón A. A new computational approach, based on images trajectories, to identify the subjacent heterogeneity of sperm to the effects of ketanserin. Cytometry A 2023; 103:655-663. [PMID: 36974731 DOI: 10.1002/cyto.a.24732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/20/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
The identification of kinematic subpopulations is of paramount importance to understanding the biological nature of the sperm heterogeneity. Nowadays, the data of motility parameters obtained by a computer-assisted sperm analysis (CASA) system has been used as input to distinct algorithms to identify kinematic subpopulations. In contrast, the images of the trajectories were depicted only as examples of the patterns of motility in each subpopulation. Here, python code was written to reconstruct the images of trajectories, from their coordinates, then the images of trajectories were used as input to a machine learning clustering algorithm of classification, and the subpopulations were described statistically by the motility parameters. Finally, the images of trajectories in each subpopulation were displayed in a way we called Pollock plots. Semen samples of boar sperm were treated with distinct concentrations of ketanserin (an antagonist of the 5-HT2 receptor of serotonin) and untreated samples were used as a control. The motility of sperm in each sample was analyzed at 0 and 30 min of incubation. Six subpopulations were found. The subpopulation 2 presented the highest values of velocities at 0 or 30 min. After 30 min of incubation, the ketanserin increased the values of the curvilinear velocity at high concentrations, whereas the linearity and the straight velocity decreased. Our computational model permits better identification of the kinematic subpopulations than the traditional approach and provides insights onto the heterogeneity of the response to ketanserin; thus, it could significantly impact the research on the relationship between sperm heterogeneity-fertility.
Collapse
Affiliation(s)
| | - Cindy U Rivas
- Laboratorio de Gametos y Desarrollo Tecnológico, Facultad de Estudios Superiores Iztacala, UNAM, 54090, Tlalnepantla, Estado de México, Mexico
| | - María E Ayala
- Unidad de Biología de la Reproducción, Laboratorio de pubertad, Facultad de Estudios Superiores Zaragoza, UNAM, Ciudad de México, 15000, Mexico
| | - Rodolfo Blanco-Rodríguez
- Department of Mathematics and Statistical Science, University of Idaho, Moscow, Idaho, USA
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, USA
| | - Nancy Juarez
- Laboratorio de Gametos y Desarrollo Tecnológico, Facultad de Estudios Superiores Iztacala, UNAM, 54090, Tlalnepantla, Estado de México, Mexico
| | - Esteban A Hernandez-Vargas
- Department of Mathematics and Statistical Science, University of Idaho, Moscow, Idaho, USA
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, USA
| | - Andrés Aragón
- Laboratorio de Gametos y Desarrollo Tecnológico, Facultad de Estudios Superiores Iztacala, UNAM, 54090, Tlalnepantla, Estado de México, Mexico
| |
Collapse
|
14
|
Cecchini K, Biasini A, Yu T, Säflund M, Mou H, Arif A, Eghbali A, Colpan C, Gainetdinov I, de Rooij DG, Weng Z, Zamore PD, Özata DM. The transcription factor TCFL5 responds to A-MYB to elaborate the male meiotic program in mice. Reproduction 2023; 165:183-196. [PMID: 36395073 PMCID: PMC9812935 DOI: 10.1530/rep-22-0355] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/17/2022] [Indexed: 11/18/2022]
Abstract
In brief The testis-specific transcription factor, TCFL5, expressed in pachytene spermatocytes regulates the meiotic gene expression program in collaboration with the transcription factor A-MYB. Abstract In male mice, the transcription factors STRA8 and MEISON initiate meiosis I. We report that STRA8/MEISON activates the transcription factors A-MYB and TCFL5, which together reprogram gene expression after spermatogonia enter into meiosis. TCFL5 promotes the transcription of genes required for meiosis, mRNA turnover, miR-34/449 production, meiotic exit, and spermiogenesis. This transcriptional architecture is conserved in rhesus macaque, suggesting TCFL5 plays a central role in meiosis and spermiogenesis in placental mammals. Tcfl5em1/em1 mutants are sterile, and spermatogenesis arrests at the mid- or late-pachytene stage of meiosis. Moreover, Tcfl5+/em1 mutants produce fewer motile sperm.
Collapse
Affiliation(s)
- Katharine Cecchini
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Adriano Biasini
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Tianxiong Yu
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Martin Säflund
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden
| | - Haiwei Mou
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Amena Arif
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
- Present address: Beam Therapeutics, 238 Main St, Cambridge, MA 02142, USA
| | - Atiyeh Eghbali
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden
| | - Cansu Colpan
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
- Present address: Voyager Therapeutics, 75 Sidney St, Cambridge, MA 02139, USA
| | - Ildar Gainetdinov
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Dirk G. de Rooij
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584, the Netherlands
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Phillip D. Zamore
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Deniz M. Özata
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden
- Lead contact
| |
Collapse
|
15
|
Morohoshi A, Miyata H, Tokuhiro K, Iida-Norita R, Noda T, Fujihara Y, Ikawa M. Testis-enriched ferlin, FER1L5, is required for Ca 2+-activated acrosome reaction and male fertility. SCIENCE ADVANCES 2023; 9:eade7607. [PMID: 36696506 PMCID: PMC9876558 DOI: 10.1126/sciadv.ade7607] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/23/2022] [Indexed: 05/28/2023]
Abstract
Spermatozoa need to undergo an exocytotic event called the acrosome reaction before fusing with eggs. Although calcium ion (Ca2+) is essential for the acrosome reaction, its molecular mechanism remains unknown. Ferlin is a single transmembrane protein with multiple Ca2+-binding C2 domains, and there are six ferlins, dysferlin (DYSF), otoferlin (OTOF), myoferlin (MYOF), fer-1-like 4 (FER1L4), FER1L5, and FER1L6, in mammals. Dysf, Otof, and Myof knockout mice have been generated, and each knockout mouse line exhibited membrane fusion disorders such as muscular dystrophy in Dysf, deafness in Otof, and abnormal myogenesis in Myof. Here, by generating mutant mice of Fer1l4, Fer1l5, and Fer1l6, we found that only Fer1l5 is required for male fertility. Fer1l5 mutant spermatozoa could migrate in the female reproductive tract and reach eggs, but no acrosome reaction took place. Even a Ca2+ ionophore cannot induce the acrosome reaction in Fer1l5 mutant spermatozoa. These results suggest that FER1L5 is the missing link between Ca2+ and the acrosome reaction.
Collapse
Affiliation(s)
- Akane Morohoshi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka 5650871 Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
| | - Keizo Tokuhiro
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 5731191 Japan
| | - Rie Iida-Norita
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
| | - Taichi Noda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Kumamoto 8600811 Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto, Kumamoto 8608555 Japan
| | - Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Osaka 5648565, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871 Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka 5650871 Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 1088639 Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka 5650871 Japan
| |
Collapse
|
16
|
Tourmente M, Sansegundo E, Rial E, Roldan ERS. Bioenergetic changes in response to sperm capacitation and two-way metabolic compensation in a new murine model. Cell Mol Life Sci 2023; 80:11. [PMID: 36534181 PMCID: PMC9763147 DOI: 10.1007/s00018-022-04652-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/29/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022]
Abstract
The acquisition of fertilizing ability by mammalian spermatozoa, known as "capacitation," includes processes that depend on particular metabolic pathways. This has led to the hypothesis that ATP demands might differ between capacitated and non-capacitated cells. Mouse sperm can produce ATP via OXPHOS and aerobic glycolysis, an advantageous characteristic considering that these cells have to function in the complex and variable environment of the female reproductive tract. Nonetheless, despite evidence showing that both metabolic pathways play a role in events associated with mouse sperm capacitation, there is contradictory evidence regarding changes promoted by capacitation in this species. In addition, the vast majority of studies regarding murine sperm metabolism use Mus musculus laboratory strains as model, thus neglecting the wide diversity of sperm traits of other species of Mus. Focus on closely related species with distinct evolutionary histories, which may be the result of different selective pressures, could shed light on diversity of metabolic processes. Here, we analyzed variations in sperm bioenergetics associated with capacitation in spermatozoa of the steppe mouse, Mus spicilegus, a species with high sperm performance. Furthermore, we compared sperm metabolic traits of this species with similar traits previously characterized in M. musculus. We found that the metabolism of M. spicilegus sperm responded to capacitation in a manner similar to that of M. musculus sperm. However, M. spicilegus sperm showed distinct metabolic features, including the ability to perform cross-pathway metabolic compensation in response to either respiratory or glycolytic inhibition, thus revealing a delicate fine-tuning of its metabolic capacities.
Collapse
Affiliation(s)
- Maximiliano Tourmente
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, CSIC, Madrid, Spain.
- Centro de Biología Celular y Molecular, Facultad de Ciencias Exactas, Físicas y Naturales (FCEFyN - UNC), Universidad Nacional de Córdoba, Córdoba, Argentina.
- Instituto de Investigaciones Biológicas y Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IIByT - CONICET, UNC), Córdoba, Argentina.
| | - Ester Sansegundo
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, CSIC, Madrid, Spain
| | - Eduardo Rial
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Eduardo R S Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, CSIC, Madrid, Spain.
| |
Collapse
|
17
|
β-Defensin 19/119 mediates sperm chemotaxis and is associated with idiopathic infertility. Cell Rep Med 2022; 3:100825. [PMID: 36513070 PMCID: PMC9797948 DOI: 10.1016/j.xcrm.2022.100825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/26/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022]
Abstract
Sperm chemotaxis is required for guiding sperm toward the egg. However, the molecular identity of physiological chemoattractant and its involvement in infertility remain elusive. Here, we identify DEFB19/119 (mouse/human orthologs) as a physiological sperm chemoattractant. The epithelia of the female reproductive tract and the cumulus-oocyte complex secrete DEFB19/119 that elicits calcium mobilization via the CatSper channel and induces sperm chemotaxis in capacitated sperm. Manipulating the level of DEFB19 in mice determines the number of sperm arriving at the fertilization site. Importantly, we identify exon mutations in the DEFB119 gene in idiopathic infertile women with low level of DEFB119 in the follicular fluid. The level of DEFB119 correlates with the chemotactic potency of follicular fluid and predicts the infertile outcome with positive correlation. This study reveals the pivotal role of DEFB19/119 in sperm chemotaxis and demonstrates its potential application in the diagnosis of idiopathic infertility.
Collapse
|
18
|
Sanchez-Rodriguez A, Sansegundo E, Tourmente M, Roldan ERS. Effect of High Viscosity on Energy Metabolism and Kinematics of Spermatozoa from Three Mouse Species Incubated under Capacitating Conditions. Int J Mol Sci 2022; 23:ijms232315247. [PMID: 36499575 PMCID: PMC9737050 DOI: 10.3390/ijms232315247] [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: 10/30/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/07/2022] Open
Abstract
In order to sustain motility and prepare for fertilization, sperm require energy. The characterization of sperm ATP production and usage in mouse species revealed substantial differences in metabolic pathways that can be differentially affected by capacitation. Moreover, spermatozoa encounter different environments with varying viscoelastic properties in the female reproductive tract. Here, we examine whether viscosity affects sperm ATP levels and kinematics during capacitation in vitro. Sperm from three mouse species (Mus musculus, M. spretus, M. spicilegus) were incubated under capacitating conditions in a modified Tyrode's medium containing bicarbonate, glucose, pyruvate, lactate, and bovine serum albumin (mT-BH) or in a bicarbonate-free medium as a non-capacitating control. Viscosity was increased with the inclusion of polyvinylpyrrolidone. ATP was measured with a bioluminescence kit, and kinematics were examined with a computer-aided sperm analysis system. In M. musculus sperm, ATP declined during capacitation, but no differences were found between non-capacitating and capacitating sperm. In contrast, in M. spretus and M. spicilegus, ATP levels decreased in capacitating sperm. Increasing viscosity in the medium did not modify the timing or proportion of cells undergoing capacitation but did result in additional time- and concentration-dependent decreases in ATP in M. spretus and M. spicilegus under capacitating conditions. Additionally, increased viscosity altered both velocity and trajectory descriptors. The limited impact of capacitation and higher viscosity on M. musculus sperm ATP and kinematics could be related to the low intensity of postcopulatory sexual selection in this species. Responses seen in the other two species could be linked to the ability of their sperm to perform better under enhanced selective pressures.
Collapse
Affiliation(s)
- Ana Sanchez-Rodriguez
- Departmento de Biodiversidad y Biología Evolutiva, Museo Nacional de Ciencias Naturales (CSIC), 28006 Madrid, Spain
| | - Ester Sansegundo
- Departmento de Biodiversidad y Biología Evolutiva, Museo Nacional de Ciencias Naturales (CSIC), 28006 Madrid, Spain
| | - Maximiliano Tourmente
- Departmento de Biodiversidad y Biología Evolutiva, Museo Nacional de Ciencias Naturales (CSIC), 28006 Madrid, Spain
- Centro de Biología Celular y Molecular, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba (FCEFyN—UNC), Córdoba X5016GCA, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IIByT—CONICET, UNC), Córdoba X5016GCA, Argentina
| | - Eduardo R. S. Roldan
- Departmento de Biodiversidad y Biología Evolutiva, Museo Nacional de Ciencias Naturales (CSIC), 28006 Madrid, Spain
- Correspondence:
| |
Collapse
|
19
|
Sun W, Tian F, Pan H, Chang X, Xia M, Hu J, Wang Y, Li R, Li W, Yang M, Zhou Z. Flurochloridone induced abnormal spermatogenesis by damaging testicular Sertoli cells in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 246:114163. [PMID: 36240522 DOI: 10.1016/j.ecoenv.2022.114163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/09/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Flurochloridone (FLC), a selective herbicide used on a global scale, has been reported to have male reproductive toxicity whose evidence is limited, but its mechanism remains unclear. The present study was conducted to systematically explore the male reproductive toxicity of FLC, including sperm quality, spermatogenesis, toxicity targets, and potential mechanisms. METHODS Male C57BL/6 mice aged 6-7 weeks received gavage administration of FLC (365/730 mg/kg/day) for 28 consecutive days. Then, the tissue and sperm of mice were collected for analysis. We measured the gonadosomatic index and analyzed sperm concentration, motility, malformation rate, and mitochondrial membrane potential (MMP). Spermatocyte immunofluorescence staining was performed to analyze meiosis. We also performed pathological staining on the testis and epididymis tissue and TUNEL staining, immunohistochemical analysis, and ultrastructural observation on the testicular tissue. RESULTS Results showed that FLC caused testicular weight reduction, dysfunction, and architectural damage in mice, but no significant adverse effect was found in the epididymis. The exposure interfered with spermatogonial proliferation and meiosis, affecting sperm concentration, motility, kinematic parameters, morphology, and MMP, decreasing sperm quality. Furthermore, mitochondrial damage and apoptosis of testicular Sertoli cells were observed in mice treated with FLC. CONCLUSION We found that FLC has significant adverse effects on spermatogonial proliferation and meiosis. Meanwhile, apoptosis and mitochondrial damage may be the potential mechanism of Sertoli cell damage. Our study demonstrated that FLC could induce testicular Sertoli cell damage, leading to abnormal spermatogenesis, which decreased sperm quality. The data provided references for the toxicity risk and research methods of FLC application in the environment.
Collapse
Affiliation(s)
- Weiqi Sun
- School of Public Health/MOE Key Laboratory for Public Health Safety/ Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China; Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China
| | - Fang Tian
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Hongjie Pan
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Xiuli Chang
- School of Public Health/MOE Key Laboratory for Public Health Safety/ Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China
| | - Minjie Xia
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Jingying Hu
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Yuzhu Wang
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Runsheng Li
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Weihua Li
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Mingjun Yang
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China.
| | - Zhijun Zhou
- School of Public Health/MOE Key Laboratory for Public Health Safety/ Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
20
|
What is the importance of sperm subpopulations? Anim Reprod Sci 2022; 246:106844. [PMID: 34538510 DOI: 10.1016/j.anireprosci.2021.106844] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022]
Abstract
The study of sperm subpopulations spans three decades. The origin, meaning, and practical significance, however, are less clear. Current technology for assessing sperm morphology (CASA-Morph) and motility (CASA-Mot) has enabled the accurate evaluation of these features, and there are many options for data classification. Subpopulations could occur as a result of the stage of development of each spermatozoon in the subpopulation. Spermatogenesis might contribute to the production of these subpopulations. Insights from evolutionary biology and recent molecular research are indicative of the diversity among male gametes that could occur from unequal sharing of transcripts and other elements through cytoplasmic bridges between spermatids. Sperm cohorts exiting the gonads would contain different RNA and protein contents, affecting the spermatozoon physiology and associations with the surrounding environmental milieu. Subsequently, these differences could affect how spermatozoa interact with the environmental milieu (maturation, mixing with seminal plasma, and interacting with the environmental milieu, or female genital tract and female gamete). The emergence of sperm subpopulations as an outcome of evolution, related to the reproductive strategies of the species, genital tract structures, and copulatory and fertilization processes. This kind of approach in determining the importance of sperm subpopulations in fertilization capacity should have a practical impact for conducting reproductive technologies, inspiring and enabling new ways for the more efficient use of spermatozoa in the medical, animal breeding, and conservation fields. This manuscript is a contribution to the Special Issue in memory of Dr. Duane Garner.
Collapse
|
21
|
Tourmente M, Sansegundo E, Rial E, Roldan ERS. Capacitation promotes a shift in energy metabolism in murine sperm. Front Cell Dev Biol 2022; 10:950979. [PMID: 36081906 PMCID: PMC9445201 DOI: 10.3389/fcell.2022.950979] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
In mammals, sperm acquire fertilization ability after a series of physiological and biochemical changes, collectively known as capacitation, that occur inside the female reproductive tract. In addition to other requirements, sperm bioenergetic metabolism has been identified as a fundamental component in the acquisition of capacitation. Mammalian sperm produce ATP through two main metabolic processes, oxidative phosphorylation (OXPHOS) and aerobic glycolysis that are localized to two different flagellar compartments, the midpiece, and the principal piece, respectively. In mouse sperm, the occurrence of many events associated with capacitation relies on the activity of these two energy-producing pathways, leading to the hypothesis that some of these events may impose changes in sperm energetic demands. In the present study, we used extracellular flux analysis to evaluate changes in glycolytic and respiratory parameters of murine sperm that occur as a consequence of capacitation. Furthermore, we examined whether these variations affect sperm ATP sustainability. Our results show that capacitation promotes a shift in the usage ratio of the two main metabolic pathways, from oxidative to glycolytic. However, this metabolic rewiring does not seem to affect the rate at which the sperm consume ATP. We conclude that the probable function of the metabolic switch is to increase the ATP supply in the distal flagellar regions, thus sustaining the energetic demands that arise from capacitation.
Collapse
Affiliation(s)
- Maximiliano Tourmente
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, CSIC, Madrid, Spain
- Centro de Biología Celular y Molecular, Facultad de Ciencias Exactas, Físicas y Naturales (FCEFyN—UNC), Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (IIByT—CONICET, UNC), Córdoba, Argentina
- *Correspondence: Maximiliano Tourmente, ; Eduardo R. S. Roldan,
| | - Ester Sansegundo
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, CSIC, Madrid, Spain
| | - Eduardo Rial
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | - Eduardo R. S. Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, CSIC, Madrid, Spain
- *Correspondence: Maximiliano Tourmente, ; Eduardo R. S. Roldan,
| |
Collapse
|
22
|
Merges GE, Meier J, Schneider S, Kruse A, Fröbius AC, Kirfel G, Steger K, Arévalo L, Schorle H. Loss of Prm1 leads to defective chromatin protamination, impaired PRM2 processing, reduced sperm motility and subfertility in male mice. Development 2022; 149:275502. [PMID: 35608054 PMCID: PMC9270976 DOI: 10.1242/dev.200330] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 05/12/2022] [Indexed: 12/12/2022]
Abstract
One of the key events during spermiogenesis is the hypercondensation of chromatin by substitution of the majority of histones by protamines. In humans and mice, protamine 1 (PRM1/Prm1) and protamine 2 (PRM2/Prm2) are expressed in a species-specific ratio. Using CRISPR-Cas9-mediated gene editing, we generated Prm1-deficient mice and demonstrated that Prm1+/- mice were subfertile, whereas Prm1-/- mice were infertile. Prm1-/- and Prm2-/- sperm showed high levels of reactive oxygen species-mediated DNA damage and increased histone retention. In contrast, Prm1+/- sperm displayed only moderate DNA damage. The majority of Prm1+/- sperm were CMA3 positive, indicating protamine-deficient chromatin, although this was not the result of increased histone retention in Prm1+/- sperm. However, sperm from Prm1+/- and Prm1-/- mice contained high levels of incompletely processed PRM2. Furthermore, the PRM1:PRM2 ratio was skewed from 1:2 in wild type to 1:5 in Prm1+/- animals. Our results reveal that PRM1 is required for proper PRM2 processing to produce mature PRM2, which, together with PRM1, is able to hypercondense DNA. Thus, the species-specific PRM1:PRM2 ratio has to be precisely controlled in order to retain full fertility.
Collapse
Affiliation(s)
- Gina Esther Merges
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Julia Meier
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Simon Schneider
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Alexander Kruse
- Department of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Biomedical Research Center of the Justus-Liebig University, 35392 Giessen, Germany
| | - Andreas Christian Fröbius
- Department of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Biomedical Research Center of the Justus-Liebig University, 35392 Giessen, Germany
| | - Gregor Kirfel
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Klaus Steger
- Department of Urology, Pediatric Urology and Andrology, Section Molecular Andrology, Biomedical Research Center of the Justus-Liebig University, 35392 Giessen, Germany
| | - Lena Arévalo
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Hubert Schorle
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
23
|
Noda T, Blaha A, Fujihara Y, Gert KR, Emori C, Deneke VE, Oura S, Panser K, Lu Y, Berent S, Kodani M, Cabrera-Quio LE, Pauli A, Ikawa M. Sperm membrane proteins DCST1 and DCST2 are required for sperm-egg interaction in mice and fish. Commun Biol 2022; 5:332. [PMID: 35393517 PMCID: PMC8989947 DOI: 10.1038/s42003-022-03289-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 03/17/2022] [Indexed: 12/28/2022] Open
Abstract
The process of sperm-egg fusion is critical for successful fertilization, yet the underlying mechanisms that regulate these steps have remained unclear in vertebrates. Here, we show that both mouse and zebrafish DCST1 and DCST2 are necessary in sperm to fertilize the egg, similar to their orthologs SPE-42 and SPE-49 in C. elegans and Sneaky in D. melanogaster. Mouse Dcst1 and Dcst2 single knockout (KO) sperm are able to undergo the acrosome reaction and show normal relocalization of IZUMO1, an essential factor for sperm-egg fusion, to the equatorial segment. While both single KO sperm can bind to the oolemma, they show the fusion defect, resulting that Dcst1 KO males become almost sterile and Dcst2 KO males become sterile. Similar to mice, zebrafish dcst1 KO males are subfertile and dcst2 and dcst1/2 double KO males are sterile. Zebrafish dcst1/2 KO sperm are motile and can approach the egg, but are defective in binding to the oolemma. Furthermore, we find that DCST1 and DCST2 interact with each other and are interdependent. These data demonstrate that DCST1/2 are essential for male fertility in two vertebrate species, highlighting their crucial role as conserved factors in fertilization.
Collapse
Affiliation(s)
- Taichi Noda
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan
| | - Andreas Blaha
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and the Medical University of Vienna, 1030, Vienna, Austria
| | - Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Krista R Gert
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and the Medical University of Vienna, 1030, Vienna, Austria
| | - Chihiro Emori
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Victoria E Deneke
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Seiya Oura
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Karin Panser
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Yonggang Lu
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Sara Berent
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Mayo Kodani
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Luis Enrique Cabrera-Quio
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and the Medical University of Vienna, 1030, Vienna, Austria
| | - Andrea Pauli
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria.
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
24
|
Sansegundo E, Tourmente M, Roldan ERS. Energy Metabolism and Hyperactivation of Spermatozoa from Three Mouse Species under Capacitating Conditions. Cells 2022; 11:220. [PMID: 35053337 PMCID: PMC8773617 DOI: 10.3390/cells11020220] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Mammalian sperm differ widely in sperm morphology, and several explanations have been presented to account for this diversity. Less is known about variation in sperm physiology and cellular processes that can give sperm cells an advantage when competing to fertilize oocytes. Capacitation of spermatozoa, a process essential for mammalian fertilization, correlates with changes in motility that result in a characteristic swimming pattern known as hyperactivation. Previous studies revealed that sperm motility and velocity depend on the amount of ATP available and, therefore, changes in sperm movement occurring during capacitation and hyperactivation may involve changes in sperm bioenergetics. Here, we examine differences in ATP levels of sperm from three mouse species (genus Mus), differing in sperm competition levels, incubated under non-capacitating and capacitating conditions, to analyse relationships between energetics, capacitation, and swimming patterns. We found that, in general terms, the amount of sperm ATP decreased more rapidly under capacitating conditions. This descent was related to the development of a hyperactivated pattern of movement in two species (M. musculus and M. spicilegus) but not in the other (M. spretus), suggesting that, in the latter, temporal dynamics and energetic demands of capacitation and hyperactivation may be decoupled or that the hyperactivation pattern differs. The decrease in ATP levels during capacitation was steeper in species with higher levels of sperm competition than in those with lower levels. Our results suggest that, during capacitation, sperm consume more ATP than under non-capacitating conditions. This higher ATP consumption may be linked to higher velocity and lateral head displacement, which are associated with hyperactivated motility.
Collapse
Affiliation(s)
- Ester Sansegundo
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, Spanish Research Council (CSIC), 28006 Madrid, Spain;
| | - Maximiliano Tourmente
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, Spanish Research Council (CSIC), 28006 Madrid, Spain;
- Centro de Biología Celular y Molecular, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Cordoba X5016GCA, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), Consejo Nacional de Investigaciones Científica y Técnicas (CONICET), Cordoba X5016GCA, Argentina
| | - Eduardo R. S. Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, Spanish Research Council (CSIC), 28006 Madrid, Spain;
| |
Collapse
|
25
|
Boersma A, Primus J, Wagner B, Broukal V, Andersen L, Pachner B, Dahlhoff M, Rülicke T, Auer KE. Influence of sperm cryopreservation on sperm motility and proAKAP4 concentration in mice. Reprod Med Biol 2022; 21:e12480. [PMID: 35919386 PMCID: PMC9336535 DOI: 10.1002/rmb2.12480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/13/2022] [Accepted: 07/04/2022] [Indexed: 02/06/2023] Open
Abstract
Background The protein proAKAP4 is crucial for sperm motility and has been suggested as an indicator of male fertility. We determined the relationship between proAKAP4 concentration and sperm motility parameters in mice, and investigated the effects of cryopreservation on these variables. Methods Computer-assisted sperm analysis and ELISA were applied to determine sperm motility and proAKAP4 concentration in fresh and frozen-thawed epididymal sperm of SWISS, B6D2F1, C57BL/6N, and BALB/c mice. Results ProAKAP4 levels ranged between 12 and 89 ng/ml and did not differ between fresh and frozen-thawed samples, or between strains. We found a negative relationship between proAKAP4 levels and some sperm motility parameters. Sperm traits differed between strains, and cryopreservation negatively affected sperm velocity but not sperm direction parameters. Conclusion ProAKAP4 levels in epididymal mouse spermatozoa were unaffected by cryopreservation, highlighting the robustness of this parameter as a potentially time-independent marker for sperm motility and fertility. The high individual variation in proAKAP4 levels supports the potential role of proAKAP4 as a marker for sperm quality, though we found no positive, and even negative relationships between proAKAP4 levels and some sperm motility parameters. Future studies have to investigate the significance of proAKAP4 as an indicator for fertility in mice.
Collapse
Affiliation(s)
- Auke Boersma
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Jasmin Primus
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Bettina Wagner
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Veronika Broukal
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
- Department of RadiologyCharité – Universitätsmedizin BerlinBerlinGermany
| | - Lill Andersen
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Barbara Pachner
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Maik Dahlhoff
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Thomas Rülicke
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Kerstin E. Auer
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| |
Collapse
|
26
|
Ma X, Wang B, Li Z, Ding X, Wen Y, Shan W, Hu W, Wang X, Xia Y. Effects of glufosinate-ammonium on male reproductive health: Focus on epigenome and transcriptome in mouse sperm. CHEMOSPHERE 2022; 287:132395. [PMID: 34597628 DOI: 10.1016/j.chemosphere.2021.132395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/18/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023]
Abstract
Glufosinate-ammonium (GLA) is a widely used herbicide with emerging concern over its neural and reproductive toxicity. To uncover potential effects of GLA on male reproductive health in mammals, adult male C57BL/6J mice were administered 0.2 mg/kg·d GLA for 5 weeks. After examination on fertility, testis histology and semen quality in the GLA group, we performed deep sequencing to identify repressive epigenetic marks including DNA methylation and histone modifications (H3K27me3 and H3K9me3), together with mRNA transcript levels in sperm. Then, we integrated multi-omics sequencing data to comprehensively explore GLA-induced epigenetic and transcriptomic alterations. We found no significant difference either on fertility, testis histology or semen quality-related indicators. As for epigenome, the protein level of H3K27me3 was significantly increased in GLA sperm. Next generation sequencing showed alterations of these epigenetic marks and extensive transcription inhibition in sperm. These differential repressive marks were mainly distributed at intergenic regions and introns. According to results by Gene Ontology enrichment analysis, both differentially methylated and expressed genes were mainly enriched in pathways related to synapse organization. Subtle differences in genomic imprinting were also observed between the two groups. These results suggested that GLA predominantly impaired sperm epigenome and transcriptome in mice, with little effect on fertility, testis histology or semen quality. Further studies on human sperm using similar strategies need to be conducted for a better understanding of the male reproductive toxicity of GLA.
Collapse
Affiliation(s)
- Xuan Ma
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Bingqian Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhe Li
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xingwang Ding
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Ya Wen
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wenqi Shan
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Weiyue Hu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
27
|
Silva AAS, Raimundo TRF, Mariani NAP, Kushima H, Avellar MCW, Buffone MG, Paula-Lopes FF, Moura MT, Silva EJR. Dissecting EPPIN protease inhibitor domains in sperm motility and fertilizing ability: repercussions for male contraceptive development. Mol Hum Reprod 2021; 27:gaab066. [PMID: 34792600 DOI: 10.1093/molehr/gaab066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/28/2021] [Indexed: 11/14/2022] Open
Abstract
EPPIN (epididymal protease inhibitor) is a mammalian conserved sperm-binding protein displaying an N-terminal WFDC (whey-acidic protein four-disulfide core) and a C-terminal Kunitz protease inhibitor domains. EPPIN plays a key role in regulating sperm motility after ejaculation via interaction with the seminal plasma protein SEMG1 (semenogelin-1). EPPIN ligands targeting the SEMG1 binding site in the Kunitz domain are under development as male contraceptive drugs. Nevertheless, the relative contributions of EPPIN WFDC and Kunitz domains to sperm function remain obscure. Here, we evaluated the effects of antibodies targeting specific epitopes in EPPIN's WFDC (Q20E antibody, Gln20-Glu39 epitope) and Kunitz (S21C and F21C antibodies, Ser103-Cys123 and Phe90-C110 epitopes, respectively) domains on mouse sperm motility and fertilizing ability. Computer-assisted sperm analysis showed that sperm co-incubation with S21C antibody (but not F21C antibody) lowered progressive and hyperactivated motilities and impaired kinematic parameters describing progressive (straight-line velocity; VSL, average path velocity; VAP and straightness; STR) and vigorous sperm movements (curvilinear velocity; VCL, amplitude of lateral head movement; ALH, and linearity; LIN) compared with control. Conversely, Q20E antibody-induced milder inhibition of progressive motility and kinematic parameters (VAP, VCL and ALH). Sperm co-incubation with S21C or Q20E antibodies affected in vitro fertilization as revealed by reduced cleavage rates, albeit without changes in capacitation-induced tyrosine phosphorylation. In conclusion, we show that targeting specific epitopes in EPPIN Kunitz and WFDC domains inhibits sperm motility and capacitation-associated events, which decrease their fertilizing ability; nevertheless, similar observations in vivo remain to be demonstrated. Simultaneously targeting residues in S21C and Q20E epitopes is a promising approach for the rational design of EPPIN-based ligands with spermostatic activity.
Collapse
Affiliation(s)
- Alan A S Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University, Botucatu-SP, Brazil
| | - Tamiris R F Raimundo
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University, Botucatu-SP, Brazil
| | - Noemia A P Mariani
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University, Botucatu-SP, Brazil
| | - Hélio Kushima
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University, Botucatu-SP, Brazil
| | - Maria Christina W Avellar
- Department of Pharmacology, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo-SP, Brazil
| | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Fabíola F Paula-Lopes
- Department of Biological Sciences, Universidade Federal de São Paulo-Campus Diadema, Diadema-SP, Brazil
| | - Marcelo T Moura
- Department of Biological Sciences, Universidade Federal de São Paulo-Campus Diadema, Diadema-SP, Brazil
| | - Erick J R Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University, Botucatu-SP, Brazil
| |
Collapse
|
28
|
Impact of Cryopreservation on Motile Subpopulations and Tyrosine-Phosphorylated Regions of Ram Spermatozoa during Capacitating Conditions. BIOLOGY 2021; 10:biology10111213. [PMID: 34827206 PMCID: PMC8614982 DOI: 10.3390/biology10111213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 11/24/2022]
Abstract
Simple Summary Spermatozoa go through diverse changes to achieve their fertilizing potential (capacitation) and develop a specific motility pattern (hyperactivation). However, to ensure a greater reproductive success, not all the spermatozoa present in an ejaculate react equally or at the same time. Therefore, a comparative analysis was performed in the present study to improve our current understanding about how cryopreservation may affect the heterogeneous nature of fresh ejaculates during these two events. Among the four motile sperm subpopulations identified in fresh and frozen-thawed ram semen, one of them developed a hyperactivated-like movement and was the main group involve in those changes associated with sperm capacitation based on the marked increase and the positive correlation with mitochondrial activity and tyrosine phosphorylation, two relevant parameters that usually increase during capacitation. In addition, cryopreservation altered the distribution of the motile sperm subpopulations. Although the subpopulation with hyperactivated-like movement increased at the beginning of incubation in frozen-thawed samples, this subpopulation together with the subpopulation of rapid and progressive spermatozoa were replaced after a prolonged incubation by the subpopulation of slow spermatozoa with the lowest mitochondrial activity, which clearly indicate the reduction in sperm quality. These findings will aid to optimize the current cryopreservation and in vitro fertilization protocols. Abstract The heterogeneous nature of ejaculates highlights the relevance of studying the behavior of different sperm subpopulations. Changes in sperm motility and the increase in tyrosine phosphorylation are key events that usually occur during capacitation and can be modified by the cryopreservation process. However, the relationship between both events remains poorly defined throughout capacitation in the different sperm subpopulations. Fresh and frozen-thawed spermatozoa were incubated in capacitating (CAP) and non-capacitating (NC) media up to 240 min. Sperm kinematics, tyrosine phosphorylation and mitochondrial activity were measured by the CASA system and imaging flow cytometry. Four motile sperm subpopulations (SP) were identified in fresh and frozen-thawed ram semen after the cluster analysis. Incubation under CAP conditions over time led to greater changes in the percentage of spermatozoa included in each subpopulation compared to NC conditions, being different between fresh and frozen-thawed spermatozoa. The SP1, characterized by slow spermatozoa, progressively increased after 15 min in frozen-thawed samples incubated in both media but not in fresh ones. The SP4, characterized by fast and non-linear spermatozoa, showed a marked increase during CAP, but not under NC conditions, occurring more rapidly in frozen-thawed spermatozoa. This subpopulation (SP4) was also the only one positively and strongly correlated with mitochondrial activity and all phosphorylated sperm regions during capacitation, either in fresh or frozen-thawed samples. Our results indicated that in vitro capacitation induced significant changes in the distribution of motile sperm subpopulations, being affected by cryopreservation. Notwithstanding, the subpopulation which probably represents hyperactivated-like spermatozoa (SP4) also increased in frozen-thawed samples, occurring faster and simultaneously to the increment of mitochondrial activity and tyrosine phosphorylation of different sperm regions.
Collapse
|
29
|
Fujihara Y, Herberg S, Blaha A, Panser K, Kobayashi K, Larasati T, Novatchkova M, Theussl HC, Olszanska O, Ikawa M, Pauli A. The conserved fertility factor SPACA4/Bouncer has divergent modes of action in vertebrate fertilization. Proc Natl Acad Sci U S A 2021; 118:e2108777118. [PMID: 34556579 PMCID: PMC8488580 DOI: 10.1073/pnas.2108777118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 11/18/2022] Open
Abstract
Fertilization is the fundamental process that initiates the development of a new individual in all sexually reproducing species. Despite its importance, our understanding of the molecular players that govern mammalian sperm-egg interaction is incomplete, partly because many of the essential factors found in nonmammalian species do not have obvious mammalian homologs. We have recently identified the lymphocyte antigen-6 (Ly6)/urokinase-type plasminogen activator receptor (uPAR) protein Bouncer as an essential fertilization factor in zebrafish [S. Herberg, K. R. Gert, A. Schleiffer, A. Pauli, Science 361, 1029-1033 (2018)]. Here, we show that Bouncer's homolog in mammals, Sperm Acrosome Associated 4 (SPACA4), is also required for efficient fertilization in mice. In contrast to fish, in which Bouncer is expressed specifically in the egg, SPACA4 is expressed exclusively in the sperm. Male knockout mice are severely subfertile, and sperm lacking SPACA4 fail to fertilize wild-type eggs in vitro. Interestingly, removal of the zona pellucida rescues the fertilization defect of Spaca4-deficient sperm in vitro, indicating that SPACA4 is not required for the interaction of sperm and the oolemma but rather of sperm and the zona pellucida. Our work identifies SPACA4 as an important sperm protein necessary for zona pellucida penetration during mammalian fertilization.
Collapse
Affiliation(s)
- Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita 564-8565, Japan
| | - Sarah Herberg
- Research Institute of Molecular Pathology, Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Andreas Blaha
- Research Institute of Molecular Pathology, Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, 1030 Vienna, Austria
| | - Karin Panser
- Research Institute of Molecular Pathology, Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Kiyonori Kobayashi
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Tamara Larasati
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Maria Novatchkova
- Research Institute of Molecular Pathology, Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Hans-Christian Theussl
- Research Institute of Molecular Pathology, Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Olga Olszanska
- Research Institute of Molecular Pathology, Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan;
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Andrea Pauli
- Research Institute of Molecular Pathology, Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria;
| |
Collapse
|
30
|
Lester WC, Johnson T, Hale B, Serra N, Elgart B, Wang R, Geyer CB, Sperry AO. Aurora a kinase (AURKA) is required for male germline maintenance and regulates sperm motility in the mouse. Biol Reprod 2021; 105:1603-1616. [PMID: 34518881 DOI: 10.1093/biolre/ioab168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/12/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Aurora A kinase (AURKA) is an important regulator of cell division and is required for assembly of the mitotic spindle. We recently reported the unusual finding that this mitotic kinase is also found on the sperm flagellum. To determine its requirement in spermatogenesis, we generated conditional knockout animals with deletion of the Aurka gene in either spermatogonia or spermatocytes to assess its role in mitotic and postmitotic cells, respectively. Deletion of Aurka in spermatogonia resulted in disappearance of all developing germ cells in the testis, as expected given its vital role in mitotic cell division. Deletion of Aurka in spermatocytes reduced testis size, sperm count, and fertility, indicating disruption of meiosis or an effect on spermiogenesis in developing mice. Interestingly, deletion of Aurka in spermatocytes increased apoptosis in spermatocytes along with an increase in the percentage of sperm with abnormal morphology. Despite the increase in abnormal sperm, sperm from spermatocyte Aurka knockout mice displayed increased progressive motility. In addition, sperm lysate prepared from Aurka knockout animals had decreased protein phosphatase 1 (PP1) activity. Together, our results show that AURKA plays multiple roles in spermatogenesis, from mitotic divisions of spermatogonia to sperm morphology and motility.
Collapse
Affiliation(s)
- William C Lester
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| | - Taylor Johnson
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Ben Hale
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Nicholas Serra
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Brian Elgart
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| | - Rong Wang
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Ann O Sperry
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| |
Collapse
|
31
|
Takei GL, Tourzani DA, Paudel B, Visconti PE. Activation of cAMP-dependent phosphorylation pathways is independent of ROS production during mouse sperm capacitation. Mol Reprod Dev 2021; 88:544-557. [PMID: 34318548 DOI: 10.1002/mrd.23524] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022]
Abstract
Mammalian sperm have to undergo capacitation to fertilize the egg. At the molecular level, capacitation involves cAMP synthesis, protein kinase A activation, and downstream increase in tyrosine phosphorylation. In addition, during capacitation, mammalian sperm actively generate reactive oxygen species (ROS). It has been proposed that ROS modulate phosphorylation pathways; however, the crosstalk between these signaling processes is not well-understood. In the present study, we used loss- and gain-of-function approaches to evaluate the interconnection between ROS and phosphorylation. We showed that BSA and HCO3 - , but not Ca2+ , in the capacitation media are required for ROS production. The synergic effect of these compounds was neither mediated by HCO3 - stimulation of cAMP synthesis nor by BSA-induced cholesterol efflux. The capacitation-induced ROS generation was blocked in the presence of superoxide dismutase (SOD), catalase, and apocynin. However, none of these compounds affected cAMP-dependent or tyrosine phosphorylation. On the other hand, the addition of NADPH to the media induced ROS generation in sperm incubated in the absence of BSA and HCO3 - without upregulating cAMP-dependent or tyrosine phosphorylation signaling. Most interestingly, catalase, but not SOD, blocked in vitro fertilization suggesting a role for H2 O2 in this process.
Collapse
Affiliation(s)
- Gen L Takei
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Darya A Tourzani
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Bidur Paudel
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
32
|
You JB, McCallum C, Wang Y, Riordon J, Nosrati R, Sinton D. Machine learning for sperm selection. Nat Rev Urol 2021; 18:387-403. [PMID: 34002070 DOI: 10.1038/s41585-021-00465-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 02/04/2023]
Abstract
Infertility rates and the number of couples seeking fertility care have increased worldwide over the past few decades. Over 2.5 million cycles of assisted reproductive technologies are being performed globally every year, but the success rate has remained at ~33%. Machine learning, an automated method of data analysis based on patterns and inference, is increasingly being deployed within the health-care sector to improve diagnostics and therapeutics. This technique is already aiding embryo selection in some fertility clinics, and has also been applied in research laboratories to improve sperm analysis and selection. Tremendous opportunities exist for machine learning to advance male fertility treatments. The fundamental challenge of sperm selection - selecting the most promising candidate from 108 gametes - presents a challenge that is uniquely well-suited to the high-throughput capabilities of machine learning algorithms paired with modern data processing capabilities.
Collapse
Affiliation(s)
- Jae Bem You
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada.,Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Christopher McCallum
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Yihe Wang
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Jason Riordon
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Reza Nosrati
- Department of Mechanical & Aerospace Engineering, Monash University, Clayton, VIC, Australia
| | - David Sinton
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
33
|
ARMC12 regulates spatiotemporal mitochondrial dynamics during spermiogenesis and is required for male fertility. Proc Natl Acad Sci U S A 2021; 118:2018355118. [PMID: 33536340 PMCID: PMC8017931 DOI: 10.1073/pnas.2018355118] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Although formation of the mitochondrial sheath is a critical process in the formation of mature spermatozoa, the molecular mechanisms involved in mitochondrial sheath genesis remain unclear. Using gene-manipulated mice, we discovered that ARMC12 regulates spatiotemporal “sperm mitochondrial dynamics” during mitochondrial sheath formation through interactions with mitochondrial proteins MIC60, VDAC2, and VDAC3 as well as testis-specific proteins TBC1D21 and GK2. In addition, we demonstrated that ARMC12-interacting proteins TBC1D21 and GK2 are also essential for mitochondrial sheath formation. Our paper sheds light on the molecular mechanisms of mitochondrial sheath formation and the regulation of sperm mitochondrial dynamics, allowing us to further understand the biology of spermatogenesis and the etiology of infertility in men. The mammalian sperm midpiece has a unique double-helical structure called the mitochondrial sheath that wraps tightly around the axoneme. Despite the remarkable organization of the mitochondrial sheath, the molecular mechanisms involved in mitochondrial sheath formation are unclear. In the process of screening testis-enriched genes for functions in mice, we identified armadillo repeat-containing 12 (ARMC12) as an essential protein for mitochondrial sheath formation. Here, we engineered Armc12-null mice, FLAG-tagged Armc12 knock-in mice, and TBC1 domain family member 21 (Tbc1d21)-null mice to define the functions of ARMC12 in mitochondrial sheath formation in vivo. We discovered that absence of ARMC12 causes abnormal mitochondrial coiling along the flagellum, resulting in reduced sperm motility and male sterility. During spermiogenesis, sperm mitochondria in Armc12-null mice cannot elongate properly at the mitochondrial interlocking step which disrupts abnormal mitochondrial coiling. ARMC12 is a mitochondrial peripheral membrane protein and functions as an adherence factor between mitochondria in cultured cells. ARMC12 in testicular germ cells interacts with mitochondrial proteins MIC60, VDAC2, and VDAC3 as well as TBC1D21 and GK2, which are required for mitochondrial sheath formation. We also observed that TBC1D21 is essential for the interaction between ARMC12 and VDAC proteins in vivo. These results indicate that ARMC12 uses integral mitochondrial membrane proteins VDAC2 and VDAC3 as scaffolds to link mitochondria and works cooperatively with TBC1D21. Thus, our studies have revealed that ARMC12 regulates spatiotemporal mitochondrial dynamics to form the mitochondrial sheath through cooperative interactions with several proteins on the sperm mitochondrial surface.
Collapse
|
34
|
Foot NJ, Gonzalez MB, Gembus K, Fonseka P, Sandow JJ, Nguyen TT, Tran D, Webb AI, Mathivanan S, Robker RL, Kumar S. Arrdc4-dependent extracellular vesicle biogenesis is required for sperm maturation. J Extracell Vesicles 2021; 10:e12113. [PMID: 34188787 PMCID: PMC8217992 DOI: 10.1002/jev2.12113] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 01/04/2023] Open
Abstract
Extracellular vesicles (EVs) are important players in cell to cell communication in reproductive systems. Notably, EVs have been found and characterized in the male reproductive tract, however, direct functional evidence for their importance in mediating sperm function is lacking. We have previously demonstrated that Arrdc4, a member of the α-arrestin protein family, is involved in extracellular vesicle biogenesis and release. Here we show that Arrdc4-mediated extracellular vesicle biogenesis is required for proper sperm function. Sperm from Arrdc4-/- mice develop normally through the testis but fail to acquire adequate motility and fertilization capabilities through the epididymis, as observed by reduced motility, premature acrosome reaction, reduction in zona pellucida binding and two-cell embryo production. We found a significant reduction in extracellular vesicle production by Arrdc4-/- epididymal epithelial cells, and further, supplementation of Arrdc4-/- sperm with additional vesicles dampened the acrosome reaction defect and restored zona pellucida binding. These results indicate that Arrdc4 is important for proper sperm maturation through the control of extracellular vesicle biogenesis.
Collapse
Affiliation(s)
- Natalie J. Foot
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth AustraliaAustralia
- School of MedicineRobinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Macarena B. Gonzalez
- School of MedicineRobinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Kelly Gembus
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth AustraliaAustralia
| | - Pamali Fonseka
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular SciencesLa Trobe UniversityMelbourneVictoriaAustralia
| | - Jarrod J. Sandow
- Advanced Technology and Biology DivisionWalter and Eliza Hall InstituteParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
| | - Thuy Tien Nguyen
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth AustraliaAustralia
- School of Biological SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Diana Tran
- School of Chemical Engineering & Advanced MaterialsUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Andrew I. Webb
- Advanced Technology and Biology DivisionWalter and Eliza Hall InstituteParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
| | - Suresh Mathivanan
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular SciencesLa Trobe UniversityMelbourneVictoriaAustralia
| | - Rebecca L. Robker
- School of MedicineRobinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Department of Anatomy and Developmental BiologyBiomedicine Discovery InstituteMonash UniversityMelbourneVictoriaAustralia
| | - Sharad Kumar
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth AustraliaAustralia
- Faculty of Health and Medical SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
| |
Collapse
|
35
|
Wang F, Gervasi MG, Bošković A, Sun F, Rinaldi VD, Yu J, Wallingford MC, Tourzani DA, Mager J, Zhu LJ, Rando OJ, Visconti PE, Strittmatter L, Bach I. Deficient spermiogenesis in mice lacking Rlim. eLife 2021; 10:e63556. [PMID: 33620316 PMCID: PMC7935487 DOI: 10.7554/elife.63556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
The X-linked gene Rlim plays major roles in female mouse development and reproduction, where it is crucial for the maintenance of imprinted X chromosome inactivation in extraembryonic tissues of embryos. However, while females carrying a systemic Rlim knockout (KO) die around implantation, male Rlim KO mice appear healthy and are fertile. Here, we report an important role for Rlim in testis where it is highly expressed in post-meiotic round spermatids as well as in Sertoli cells. Systemic deletion of the Rlim gene results in lower numbers of mature sperm that contains excess cytoplasm, leading to decreased sperm motility and in vitro fertilization rates. Targeting the conditional Rlim cKO specifically to the spermatogenic cell lineage largely recapitulates this phenotype. These results reveal functions of Rlim in male reproduction specifically in round spermatids during spermiogenesis.
Collapse
Affiliation(s)
- Feng Wang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Maria Gracia Gervasi
- Department of Veterinary & Animal Sciences, University of Massachusetts AmherstAmherstUnited States
| | - Ana Bošković
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Fengyun Sun
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Vera D Rinaldi
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Jun Yu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Mary C Wallingford
- Department of Veterinary & Animal Sciences, University of Massachusetts AmherstAmherstUnited States
| | - Darya A Tourzani
- Department of Veterinary & Animal Sciences, University of Massachusetts AmherstAmherstUnited States
| | - Jesse Mager
- Department of Veterinary & Animal Sciences, University of Massachusetts AmherstAmherstUnited States
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical SchoolWorcesterUnited States
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Oliver J Rando
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Pablo E Visconti
- Department of Veterinary & Animal Sciences, University of Massachusetts AmherstAmherstUnited States
| | - Lara Strittmatter
- Electron Microscopy Core, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Ingolf Bach
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical SchoolWorcesterUnited States
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| |
Collapse
|
36
|
Alagbonsi AI, Olayaki LA. Vitamin C ameliorates tetrahydrocannabinol-induced spermatotoxicity in-vitro. BMC Nutr 2020; 6:59. [PMID: 33292756 PMCID: PMC7684963 DOI: 10.1186/s40795-020-00387-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/30/2020] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND We investigated the in-vitro effects of vitamin C on delta-9-tetrahydrocannabinol (THC) -induced reduction in spermatozoa motility and kinematics. METHODS Six rats were used for the study. Semen from each of the 6 rats was randomly divided into 6 groups such that each rat's semen was in all of the groups. Groups I-III received placebo, THC (1 mM), and vitamin C (5 mM) respectively. Group IV was pre-treated with cannabinoid receptors' blockers (CBs-) 1 and 2, followed by THC. Groups V and VI received THC and vitamin C, but group VI was additionally pre-treated with CBs-. RESULTS The spermatozoa progressive motility, average path velocity (VAP), curvilinear velocity (VCL), straight-line velocity (VSL), amplitude of lateral head (ALH) and beat cross frequency (BCF) were reduced by THC (6.08 ± 1.16%; 5.64 ± 0.82 μm/s; 6.96 ± 0.74 μm/s; 2.75 ± 0.23 μm/s; 0.31 ± 0.02 μm; and 0.78 ± 0.08 Hz respectively) but increased by vitamin C (51.20 ± 1.32%; 17.90 ± 0.21 μm/s; 25.11 ± 0.96 μm/s; 8.80 ± 0.27 μm/s; 0.75 ± 0.01 μm; and 3.15 ± 0.03 Hz respectively) when compared to control (39.72 ± 0.38%; 13.70 ± 0.29 μm/s; 18.04 ± 0.58 μm/s; 7.54 ± 0.34 μm/s; 0.65 ± 0.02 μm; and 2.79 ± 0.01 Hz respectively). Vitamin C inhibited the THC-induced reduction in these parameters (37.36 ± 0.73%; 10.98 ± 0.45 μm/s; 13.58 ± 0.30 μm/s; 7.11 ± 0.22 μm/s; 0.58 ± 0.01 μm; and 2.60 ± 0.01 Hz respectively) in the absence of CBs- 1 and 2, and even caused additional increases in progressive motility (49.54 ± 1.01%), VAP (15.70 ± 0.38 μm/s) and VCL (22.53 ± 0.29 μm/s) above the control levels with CBs-. CONCLUSION Vitamin C ameliorates the THC-induced reduction in spermatozoa motility in-vitro by modulation of their kinematics.
Collapse
Affiliation(s)
- Abdullateef Isiaka Alagbonsi
- Department of Physiology, School of Medicine and Pharmacy, University of Rwanda College of Medicine and Health Sciences, Huye, Republic of Rwanda.
| | - Luqman Aribidesi Olayaki
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria
| |
Collapse
|
37
|
Miyamoto Y, Sasaki M, Miyata H, Monobe Y, Nagai M, Otani M, Whiley PAF, Morohoshi A, Oki S, Matsuda J, Akagi KI, Adachi J, Okabe M, Ikawa M, Yoneda Y, Loveland KL, Oka M. Genetic loss of importin α4 causes abnormal sperm morphology and impacts on male fertility in mouse. FASEB J 2020; 34:16224-16242. [PMID: 33058343 DOI: 10.1096/fj.202000768rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 11/11/2022]
Abstract
Importin α proteins play a central role in the transport of cargo from the cytoplasm to the nucleus. In this study, we observed that male knock-out mice for importin α4, which is encoded by the Kpna4 gene (Kpna4-/- ), were subfertile and yielded smaller litter sizes than those of wild-type (WT) males. In contrast, mice lacking the closely related importin α3 (Kpna3-/- ) were fertile. In vitro fertilization and sperm motility assays demonstrated that sperm from Kpna4-/- mice had significantly reduced quality and motility. In addition, acrosome reaction was also impaired in Kpna4-/- mice. Transmission electron microscopy revealed striking defects, including abnormal head morphology and multiple axoneme structures in the flagella of Kpna4-/- mice. A five-fold increase in the frequency of abnormalities in Kpna4-/- mice compared to WT mice indicates the functional importance of importin α4 in normal sperm development. Moreover, Nesprin-2, which is a component of the linker of nucleus and cytoskeleton complex, was expressed at lower levels in sperm from Kpna4-/- mice and was localized with abnormal axonemes, suggesting incorrect formation of the nuclear membrane-cytoskeleton structure during spermiogenesis. Proteomics analysis of Kpna4-/- testis showed significantly altered expression of proteins related to sperm formation, which provided evidence that genetic loss of importin α4 perturbed chromatin status. Collectively, these findings indicate that importin α4 is critical for establishing normal sperm morphology in mice, providing new insights into male germ cell development by highlighting the requirement of importin α4 for normal fertility.
Collapse
Affiliation(s)
- Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Mitsuho Sasaki
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoko Monobe
- Section of Laboratory Equipment, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Masahiro Nagai
- Department of Frontier Bioscience, Graduate School of Frontier Bioscience, Osaka University, Osaka, Japan
| | - Mayumi Otani
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Penny A F Whiley
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Akane Morohoshi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinya Oki
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junichiro Matsuda
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Ken-Ichi Akagi
- Section of Laboratory Equipment, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Jun Adachi
- Laboratory of Proteome Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Masaru Okabe
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshihiro Yoneda
- National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Kate L Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Masahiro Oka
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| |
Collapse
|
38
|
Finkelstein M, Etkovitz N, Breitbart H. Ca 2+ signaling in mammalian spermatozoa. Mol Cell Endocrinol 2020; 516:110953. [PMID: 32712383 DOI: 10.1016/j.mce.2020.110953] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 06/08/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
Calcium is an essential ion which regulates sperm motility, capacitation and the acrosome reaction (AR), three processes necessary for successful fertilization. The AR enables the spermatozoon to penetrate into the egg. In order to undergo the AR, the spermatozoon must reside in the female reproductive tract for several hours, during which a series of biochemical transformations takes place, collectively called capacitation. An early event in capacitation is relatively small elevation of intracellular Ca2+ (in the nM range) and bicarbonate, which collectively activate the soluble adenylyl cyclase to produce cyclic-AMP; c-AMP activates protein kinase A (PKA), leading to indirect tyrosine phosphorylation of proteins. During capacitation, there is an increase in the membrane-bound phospholipase C (PLC) which is activated prior to the AR by relatively high increase in intracellular Ca2+ (in the μM range). PLC catalyzes the hydrolysis of phosphatidyl-inositol-4,5-bisphosphate (PIP2) to diacylglycerol and inositol-trisphosphate (IP3), leading to activation of protein kinase C (PKC) and the IP3-receptor. PKC activates a Ca2+- channel in the plasma membrane, and IP3 activates the Ca2+- channel in the outer acrosomal membrane, leading to Ca2+ depletion from the acrosome. As a result, the plasma-membrane store-operated Ca2+ channel (SOCC) is activated to increase cytosolic Ca2+ concentration, enabling completion of the acrosome reaction. The hydrolysis of PIP2 by PLC results in the release and activation of PIP2-bound gelsolin, leading to F-actin dispersion, an essential step prior to the AR. Ca2+ is also involved in the regulation of sperm motility. During capacitation, the sperm develops a unique motility pattern called hyper-activated motility (HAM) which is essential for successful fertilization. The main Ca2+-channel that mediates HAM is the sperm-specific CatSper located in the sperm tail.
Collapse
Affiliation(s)
| | - Nir Etkovitz
- Sperm Bank, Sheba Hospital, Tel-Hashomer, Israel
| | - Haim Breitbart
- The Mina & Everard Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel.
| |
Collapse
|
39
|
Devlin DJ, Nozawa K, Ikawa M, Matzuk MM. Knockout of family with sequence similarity 170 member A (Fam170a) causes male subfertility, while Fam170b is dispensable in mice†. Biol Reprod 2020; 103:205-222. [PMID: 32588889 PMCID: PMC7401401 DOI: 10.1093/biolre/ioaa082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/09/2020] [Accepted: 05/21/2020] [Indexed: 01/08/2023] Open
Abstract
Families with sequence similarity 170 members A and B (FAM170A and FAM170B) are testis-specific, paralogous proteins that share 31% amino acid identity and are conserved throughout mammals. While previous in vitro experiments suggested that FAM170B, an acrosome-localized protein, plays a role in the mouse sperm acrosome reaction and fertilization, the role of FAM170A in the testis has not been explored. In this study, we used CRISPR/Cas9 to generate null alleles for each gene, and homozygous null (-/-) male mice were mated to wild-type females for 6 months to assess fertility. Fam170b-/- males were found to produce normal litter sizes and had normal sperm counts, motility, and sperm morphology. In contrast, mating experiments revealed significantly reduced litter sizes and a reduced pregnancy rate from Fam170a-/- males compared with controls. Fam170a-/-;Fam170b-/- double knockout males also produced markedly reduced litter sizes, although not significantly different from Fam170a-/- alone, suggesting that Fam170b does not compensate for the absence of Fam170a. Fam170a-/- males exhibited abnormal spermiation, abnormal head morphology, and reduced progressive sperm motility. Thus, FAM170A has an important role in male fertility, as the loss of the protein leads to subfertility, while FAM170B is expendable. The molecular functions of FAM170A in spermatogenesis are as yet unknown; however, the protein localizes to the nucleus of elongating spermatids and may mediate its effects on spermatid head shaping and spermiation by regulating the expression of other genes. This work provides the first described role of FAM170A in reproduction and has implications for improving human male infertility diagnoses.
Collapse
Affiliation(s)
- Darius J Devlin
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Kaori Nozawa
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Toyko, Japan
| | - Martin M Matzuk
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
40
|
Liu M, Zhu Y, Xing F, Liu S, Xia Y, Jiang Q, Qin J. The polycomb group protein PCGF6 mediates germline gene silencing by recruiting histone-modifying proteins to target gene promoters. J Biol Chem 2020; 295:9712-9724. [PMID: 32482889 PMCID: PMC7363147 DOI: 10.1074/jbc.ra119.012121] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/27/2020] [Indexed: 01/18/2023] Open
Abstract
Polycomb group (PcG) proteins are essential for maintenance of lineage fidelity by coordinating developmental gene expression programs. Polycomb group ring finger 6 (PCGF6) has been previously reported to repress expression of lineage-specific genes, especially germ cell-related genes in mouse embryonic stem cells (ESCs) via the noncanonical polycomb repressive complex PRC1.6. However, the molecular mechanism of this repression remains largely unknown. Here, using RNA-Seq, real-time RT-PCR, immunohistochemistry, immunoprecipitation, and ChIP analyses, we demonstrate that PCGF6 plays an essential role in embryonic development, indicated by the partially penetrant embryonic lethality in homozygous PCGF6 (Pcgf6-/-)-deficient mice. We also found that surviving Pcgf6-deficient mice exhibit reduced fertility. Using the Pcgf6-deficient mice, we observed that ablation of Pcgf6 in somatic tissues robustly derepresses germ cell-related genes. We further provide evidence that these genes are direct targets of PCGF6 in ESCs and that endogenous PCGF6 co-localizes with the histone-modifying proteins G9A histone methyltransferase (G9A)/G9a-like protein (GLP) and histone deacetylase 1/2 (HDAC1/2) on the promoters of the germ cell-related genes. Moreover, the binding of these proteins to their target genes correlated with methylation of Lys-9 of histone 3 and with the status of histone acetylation at these genes. Moreover, the recruitment of G9A/GLP and HDAC1/2 to target promoters depended on the binding of PCGF6. Our findings indicate that PCGF6 has a critical role in safeguarding lineage decisions and in preventing aberrant expression of germ cell-related genes.
Collapse
Affiliation(s)
- Mengjie Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yaru Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Fei Xing
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shuang Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Yin Xia
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Jinzhong Qin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China
| |
Collapse
|
41
|
Wu PH, Fu Y, Cecchini K, Özata DM, Arif A, Yu T, Colpan C, Gainetdinov I, Weng Z, Zamore PD. The evolutionarily conserved piRNA-producing locus pi6 is required for male mouse fertility. Nat Genet 2020; 52:728-739. [PMID: 32601478 PMCID: PMC7383350 DOI: 10.1038/s41588-020-0657-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/29/2020] [Indexed: 12/16/2022]
Abstract
Pachytene PIWI-interacting RNAs (piRNAs), which comprise >80% of small RNAs in the adult mouse testis, have been proposed to bind and regulate target RNAs like microRNAs, cleave targets like short interfering RNAs or lack biological function altogether. Although piRNA pathway protein mutants are male sterile, no biological function has been identified for any mammalian piRNA-producing locus. Here, we report that males lacking piRNAs from a conserved mouse pachytene piRNA locus on chromosome 6 (pi6) produce sperm with defects in capacitation and egg fertilization. Moreover, heterozygous embryos sired by pi6-/- fathers show reduced viability in utero. Molecular analyses suggest that pi6 piRNAs repress gene expression by cleaving messenger RNAs encoding proteins required for sperm function. pi6 also participates in a network of piRNA-piRNA precursor interactions that initiate piRNA production from a second piRNA locus on chromosome 10, as well as pi6 itself. Our data establish a direct role for pachytene piRNAs in spermiogenesis and embryo viability.
Collapse
Affiliation(s)
- Pei-Hsuan Wu
- Howard Hughes Medical Institute and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Yu Fu
- Bioinformatics Program, Boston University, Boston, MA, USA.,Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA.,Oncology Drug Discovery Unit, Takeda Pharmaceuticals, Cambridge, MA, USA
| | - Katharine Cecchini
- Howard Hughes Medical Institute and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Deniz M Özata
- Howard Hughes Medical Institute and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Amena Arif
- Howard Hughes Medical Institute and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Tianxiong Yu
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA.,School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cansu Colpan
- Howard Hughes Medical Institute and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ildar Gainetdinov
- Howard Hughes Medical Institute and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA. .,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Phillip D Zamore
- Howard Hughes Medical Institute and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
42
|
Noda T, Fujihara Y, Matsumura T, Oura S, Kobayashi S, Ikawa M. Seminal vesicle secretory protein 7, PATE4, is not required for sperm function but for copulatory plug formation to ensure fecundity†. Biol Reprod 2020; 100:1035-1045. [PMID: 30452524 PMCID: PMC6483057 DOI: 10.1093/biolre/ioy247] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/29/2018] [Accepted: 11/16/2018] [Indexed: 12/25/2022] Open
Abstract
Seminal vesicle secretions (SVSs), together with spermatozoa, are ejaculated into the female reproductive tract. SVS7, also known as PATE4, is one of the major SVS proteins found in the seminal vesicle, copulatory plug, and uterine fluid after copulation. Here, we generated Pate4 knockout (-/-) mice and examined the detailed function of PATE4 on male fecundity. The morphology and weight of Pate4-/- seminal vesicles were comparable to the control. Although Pate4-/- cauda epididymal spermatozoa have no overt defects during in vitro fertilization, Pate4-/- males were subfertile. We found that the copulatory plugs were smaller in the vagina of females mated with Pate4-/- males, leading to semen leakage and a decreased sperm count in the uterus. When the females mated with Pate4-/- males were immediately re-caged with Pate4+/+ males, the females had subsequent productive matings. When the cauda epididymal spermatozoa were injected into the uterus and plugged artificially [artificial insemination (AI)], Pate4-/- spermatozoa could efficiently fertilize eggs as compared to wild-type spermatozoa. We finally examined the effect of SVSs on AI, and observed no difference in fertilization rates between Pate4+/+ and Pate4-/- SVSs. In conclusion, PATE4 is a novel factor in forming the copulatory plug that inhibits sequential matings and maintains spermatozoa in the uterus to ensure male fecundity.
Collapse
Affiliation(s)
- Taichi Noda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Seiya Oura
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Sumire Kobayashi
- 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 Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.,Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| |
Collapse
|
43
|
Sakaguchi D, Miyado K, Iwamoto T, Okada H, Yoshida K, Kang W, Suzuki M, Yoshida M, Kawano N. Human Semenogelin 1 Promotes Sperm Survival in the Mouse Female Reproductive Tract. Int J Mol Sci 2020; 21:ijms21113961. [PMID: 32486486 PMCID: PMC7312897 DOI: 10.3390/ijms21113961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 01/13/2023] Open
Abstract
Semenogelin 1 (SEMG1), a main component of human seminal plasma, is a multi-functional protein involved in the regulation of sperm motility and fertility. SEMG1 is orthologous to mouse seminal vesicle secretion 2 (SVS2), required for sperm survival in the female reproductive tract after copulation; however, its in vivo function remains unclear. In this study, we addressed this issue by examining the effect of recombinant SEMG1 on intrauterine mouse sperm survival. SEMG1 caused a dose-dependent decrease in mouse sperm motility, similar to its effect on human sperm, but SVS2 had no effect on mouse sperm motility. Mouse epididymal sperm in the presence of 100 µM SEMG1, a concentration that does not affect mouse sperm motility, were injected into the mouse uterus (intrauterine insemination, IUI). IUI combined with SEMG1 significantly increased the survival rate of intrauterine mouse sperm. The effect of SEMG1 on intrauterine sperm survival was comparable with that of SVS2. For clinical applications, three potentially sperm-protecting polypeptides that are easy to handle were designed from SEMG1, but their individual use was unable to mimic the ability of SEMG1. Our results indicate that SEMG1 has potential clinical applications for effective IUI and thereby for safe, simple, and effective internal fertilization.
Collapse
Affiliation(s)
- Daiki Sakaguchi
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan;
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.M.); (W.K.); (M.S.)
| | - Teruaki Iwamoto
- Division of Male Infertility, Center for Human Reproduction, Sanno Hospital, International University of Health and Welfare, Tokyo 107-0052, Japan;
| | - Hiroshi Okada
- Department of Urology, Dokkyo Medical University Saitama Medical Center, Saitama 343-8555, Japan;
| | - Kaoru Yoshida
- Faculty of Biomedical Engineering, Toin University of Yokohama, Kanagawa 225-8503, Japan;
| | - Woojin Kang
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.M.); (W.K.); (M.S.)
| | - Miki Suzuki
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; (K.M.); (W.K.); (M.S.)
| | - Manabu Yoshida
- Misaki Marine Biological Station, School of Science, the University of Tokyo, Kanagawa 238-0225, Japan
- Correspondence: (M.Y.); (N.K.)
| | - Natsuko Kawano
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan;
- Correspondence: (M.Y.); (N.K.)
| |
Collapse
|
44
|
Michaelis M, Sobczak A, Ludwig C, Marvanová H, Langhammer M, Schön J, Weitzel JM. Altered testicular cell type composition in males of two outbred mouse lines selected for high fertility. Andrology 2020; 8:1419-1427. [PMID: 32306511 DOI: 10.1111/andr.12802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 03/17/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Recently we described two outbred mouse lines which have been selected for high fertility. These mouse models doubled the number of offspring per litter. OBJECTIVES Although selected for a primarily female-trait of high fertility (increased litter size), we were interested whether also males of the fertility lines show differences within their reproductive organs. MATERIALS AND METHODS We investigated males from two outbred mouse lines which have been selected for the phenotype "high fertility" for more than 170 generations. In the present study, we analysed the testicular cell type composition by flow cytometry. We further investigated the weights of reproductive organs, histomorphometry of testis as well as studied sperm motility parameters using a thermal stress assay as well as a sperm hyperactivation assay. RESULTS Here, we describe that males of the fertility line (FL) 1 show an increased percentage of diploid cells within the testis. Flow cytometric analysis identified this enlarged cell population as Leydig cells. Testis weights were unaffected whereas the weights of seminal vesicles of FL1 and FL2 were increased compared to Ctrl bucks. FL2 males show decreased diameter of tubulus seminiferi and an enhanced spermatid/Sertoli cell index. Sperm motility parameters of FL1 and Ctrl males are initially indistinguishable but FL1 spermatozoa show a better performance in a thermal stress experiment over a 5 hours observation period. DISCUSSION These data indicate that although selected for a primarily female-trait of high fertility also males from the fertility lines are effected by defined alterations in their reproductive organs. CONCLUSION Some of these alterations are FL1-specific others are FL2-associated, indicating that different molecular strategies warrant the high-fertility phenotype on the female as well as on the male side.
Collapse
Affiliation(s)
- Marten Michaelis
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Alexander Sobczak
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Carolin Ludwig
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Hana Marvanová
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Martina Langhammer
- Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Jennifer Schön
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Joachim M Weitzel
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
45
|
Sperm proteins SOF1, TMEM95, and SPACA6 are required for sperm-oocyte fusion in mice. Proc Natl Acad Sci U S A 2020; 117:11493-11502. [PMID: 32393636 PMCID: PMC7261011 DOI: 10.1073/pnas.1922650117] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sperm-oocyte membrane fusion is one of the most important events for fertilization. So far, IZUMO1 and Fertilization Influencing Membrane Protein (FIMP) on the sperm membrane and CD9 and JUNO (IZUMO1R/FOLR4) on the oocyte membrane have been identified as fusion-required proteins. However, the molecular mechanisms for sperm-oocyte fusion are still unclear. Here, we show that testis-enriched genes, sperm-oocyte fusion required 1 (Sof1/Llcfc1/1700034O15Rik), transmembrane protein 95 (Tmem95), and sperm acrosome associated 6 (Spaca6), encode sperm proteins required for sperm-oocyte fusion in mice. These knockout (KO) spermatozoa carry IZUMO1 but cannot fuse with the oocyte plasma membrane, leading to male sterility. Transgenic mice which expressed mouse Sof1, Tmem95, and Spaca6 rescued the sterility of Sof1, Tmem95, and Spaca6 KO males, respectively. SOF1 and SPACA6 remain in acrosome-reacted spermatozoa, and SPACA6 translocates to the equatorial segment of these spermatozoa. The coexpression of SOF1, TMEM95, and SPACA6 in IZUMO1-expressing cultured cells did not enhance their ability to adhere to the oocyte membrane or allow them to fuse with oocytes. SOF1, TMEM95, and SPACA6 may function cooperatively with IZUMO1 and/or unknown fusogens in sperm-oocyte fusion.
Collapse
|
46
|
A framework for high-resolution phenotyping of candidate male infertility mutants: from human to mouse. Hum Genet 2020; 140:155-182. [PMID: 32248361 DOI: 10.1007/s00439-020-02159-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/27/2020] [Indexed: 12/18/2022]
Abstract
Male infertility is a heterogeneous condition of largely unknown etiology that affects at least 7% of men worldwide. Classical genetic approaches and emerging next-generation sequencing studies support genetic variants as a frequent cause of male infertility. Meanwhile, the barriers to transmission of this disease mean that most individual genetic cases will be rare, but because of the large percentage of the genome required for spermatogenesis, the number of distinct causal mutations is potentially large. Identifying bona fide causes of male infertility thus requires advanced filtering techniques to select for high-probability candidates, including the ability to test causality in animal models. The mouse remains the gold standard for defining the genotype-phenotype connection in male fertility. Here, we present a best practice guide consisting of (a) major points to consider when interpreting next-generation sequencing data performed on infertile men, and, (b) a systematic strategy to categorize infertility types and how they relate to human male infertility. Phenotyping infertility in mice can involve investigating the function of multiple cell types across the testis and epididymis, as well as sperm function. These findings will feed into the diagnosis and treatment of male infertility as well as male health broadly.
Collapse
|
47
|
Han T, Wang L, Tang W, Zhang Z, Khawar MB, Li G, Jiang H, Liu C. GGNBP1 ensures proper spermiogenesis in response to stress in mice. Biochem Biophys Res Commun 2020; 525:706-713. [PMID: 32139124 DOI: 10.1016/j.bbrc.2020.02.118] [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: 02/05/2020] [Accepted: 02/20/2020] [Indexed: 01/21/2023]
Abstract
GGNBP1 is known as gametogenetin protein 1 (GGN1)-interacting protein. It is specifically expressed in the mitochondria of the testis, while its functional role during spermatogenesis is still unknown. Here, we showed that the disruption of Ggnbp1 resulted in abnormal spermiogenesis in around 40% mice, while the others show no defects in the genital system. Moreover, upon treatment with low dose of bisphenol A (BPA), Ggnbp1 knockout mice were more sensitive to environmental pollutant than control mice. The treatment led to decrease in sperm motility and production of abnormal spermatozoa. These results suggest that GGNBP1 mainly ensures proper spermiogenesis in response to various stresses in male mice.
Collapse
Affiliation(s)
- Tingting Han
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100101, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lina Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenhao Tang
- Department of Urology, Peking University Third Hospital, Beijing, 100191, China
| | - Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing, 100191, China
| | - Muhammad Babar Khawar
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Guoping Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100101, China.
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing, 100191, China; Department of Andrology, Peking University Third Hospital, Beijing, 100191, China; Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing, 100191, China; Department of Human Sperm Bank, Peking University Third Hospital, Beijing, 100191, China.
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
48
|
Devlin DJ, Agrawal Zaneveld S, Nozawa K, Han X, Moye AR, Liang Q, Harnish JM, Matzuk MM, Chen R. Knockout of mouse receptor accessory protein 6 leads to sperm function and morphology defects†. Biol Reprod 2020; 102:1234-1247. [PMID: 32101290 DOI: 10.1093/biolre/ioaa024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/31/2019] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Receptor accessory protein 6 (REEP6) is a member of the REEP/Ypt-interacting protein family that we recently identified as essential for normal endoplasmic reticulum homeostasis and protein trafficking in the retina of mice and humans. Interestingly, in addition to the loss of REEP6 in our knockout (KO) mouse model recapitulating the retinal degeneration of humans with REEP6 mutations causing retinitis pigmentosa (RP), we also found that male mice are sterile. Herein, we characterize the infertility caused by loss of Reep6. Expression of both Reep6 mRNA transcripts is present in the testis; however, isoform 1 becomes overexpressed during spermiogenesis. In vitro fertilization assays reveal that Reep6 KO spermatozoa are able to bind the zona pellucida but are only able to fertilize oocytes lacking the zona pellucida. Although spermatogenesis appears normal in KO mice, cauda epididymal spermatozoa have severe motility defects and variable morphological abnormalities, including bent or absent tails. Immunofluorescent staining reveals that REEP6 expression first appears in stage IV tubules within step 15 spermatids, and REEP6 localizes to the connecting piece, midpiece, and annulus of mature spermatozoa. These data reveal an important role for REEP6 in sperm motility and morphology and is the first reported function for a REEP protein in reproductive processes. Additionally, this work identifies a new gene potentially responsible for human infertility and has implications for patients with RP harboring mutations in REEP6.
Collapse
Affiliation(s)
- Darius J Devlin
- Interdepartmental Program in Translational Biology & Molecular Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Smriti Agrawal Zaneveld
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Kaori Nozawa
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Xiao Han
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Reproductive Medical Center, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Abigail R Moye
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Qingnan Liang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jacob Michael Harnish
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Martin M Matzuk
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Rui Chen
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
49
|
Miyata H, Shimada K, Morohoshi A, Oura S, Matsumura T, Xu Z, Oyama Y, Ikawa M. Testis-enriched kinesin KIF9 is important for progressive motility in mouse spermatozoa. FASEB J 2020; 34:5389-5400. [PMID: 32072696 PMCID: PMC7136151 DOI: 10.1096/fj.201902755r] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 01/31/2023]
Abstract
Kinesin is a molecular motor that moves along microtubules. Kinesin family member 9 (KIF9) is evolutionarily conserved and expressed strongly in mouse testis. In the unicellular flagellate Chlamydomonas, KLP1 (ortholog of KIF9) is localized to the central pair microtubules of the axoneme and regulates flagellar motility. In contrast, the function of KIF9 remains unclear in mammals. Here, we mutated KIF9 in mice using the CRISPR/Cas9 system. Kif9 mutated mice exhibit impaired sperm motility and subfertility. Further analysis reveals that the flagella lacking KIF9 showed an asymmetric waveform pattern, which leads to a circular motion of spermatozoa. In spermatozoa that lack the central pair protein HYDIN, KIF9 was not detected by immunofluorescence and immunoblot analysis. These results suggest that KIF9 is associated with the central pair microtubules and regulates flagellar motility in mice.
Collapse
Affiliation(s)
- Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Akane Morohoshi
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Graduate School of Medicine, Osaka University, Suita, Japan
| | - Seiya Oura
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Zoulan Xu
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Yuki Oyama
- Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Graduate School of Medicine, Osaka University, Suita, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan.,The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Gimeno-Martos S, Casao A, Yeste M, Cebrián-Pérez JA, Muiño-Blanco T, Pérez-Pé R. Melatonin reduces cAMP-stimulated capacitation of ram spermatozoa. Reprod Fertil Dev 2019; 31:420-431. [PMID: 30209004 DOI: 10.1071/rd18087] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/07/2018] [Indexed: 12/15/2022] Open
Abstract
The presence of melatonin receptors on the surface of ram spermatozoa has led to speculation about melatonin having a role in sperm functionality. The aim of this study was to elucidate the mechanism through which melatonin regulates ram sperm capacitation induced by a cocktail containing cAMP-elevating agents. Cocktail samples capacitated in the presence of 1µM melatonin showed lower percentages of capacitated spermatozoa (chlortetracycline staining; P<0.001) together with a decrease in protein tyrosine phosphorylation (P<0.01) and lower levels of reactive oxygen species (ROS) and cAMP (P<0.05) compared with cocktail samples without the hormone. Determination of kinematic parameters, together with principal component and cluster analyses, allowed us to define four sperm subpopulations (SP). After 3h of incubation with cAMP-elevating agents, the percentages of spermatozoa belonging to SP1 (high straightness) and SP4 (less-vigorous spermatozoa with non-linear motility) increased while SP2 and SP3 (rapid spermatozoa starting hyperactivation or already hyperactivated) decreased compared with the control sample. The presence of melatonin at 100 pM and 10nM restored these subpopulations to values closer to those found in the control sample. These results indicate that melatonin at micromolar concentrations modulates ram sperm capacitation induced by cAMP-elevating agents, reducing ROS and cAMP levels, whereas at lower concentrations melatonin modifies motile sperm subpopulations. These findings warrant further studies on the potential use of melatonin for controlling capacitation in artificial insemination procedures.
Collapse
Affiliation(s)
- Silvia Gimeno-Martos
- Department of Biochemistry and Molecular and Cell Biology, Institute of Environmental Sciences of Aragón, School of Veterinary Medicine, University of Zaragoza, C/Miguel Servet 177, 50013, Zaragoza, Spain
| | - Adriana Casao
- Department of Biochemistry and Molecular and Cell Biology, Institute of Environmental Sciences of Aragón, School of Veterinary Medicine, University of Zaragoza, C/Miguel Servet 177, 50013, Zaragoza, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Department of Biology, Institute of Food and Agricultural Technology, University of Girona, C/Maria Aurèlia Campany 69, Campus Montilivi, E-17003 Girona, Spain
| | - José A Cebrián-Pérez
- Department of Biochemistry and Molecular and Cell Biology, Institute of Environmental Sciences of Aragón, School of Veterinary Medicine, University of Zaragoza, C/Miguel Servet 177, 50013, Zaragoza, Spain
| | - Teresa Muiño-Blanco
- Department of Biochemistry and Molecular and Cell Biology, Institute of Environmental Sciences of Aragón, School of Veterinary Medicine, University of Zaragoza, C/Miguel Servet 177, 50013, Zaragoza, Spain
| | - Rosaura Pérez-Pé
- Department of Biochemistry and Molecular and Cell Biology, Institute of Environmental Sciences of Aragón, School of Veterinary Medicine, University of Zaragoza, C/Miguel Servet 177, 50013, Zaragoza, Spain
| |
Collapse
|